You are on page 1of 528

Contemporary Diabetes

Series Editor: Aristidis Veves

Solomon Tesfaye
Christopher H. Gibbons
Rayaz Ahmed Malik
Aristidis Veves   Editors

Diabetic
Neuropathy
Advances in Pathophysiology and Clinical
Management
Third Edition
Contemporary Diabetes
Series Editor
Aristidis Veves, Beth Israel Deaconess Medical Center
Boston, MA, USA
The Contemporary Diabetes series focuses on the clinical aspects of obesity
and diabetes and provides the practicing health provider with all the latest
information regarding their management. The series also targets both basic
and clinical researchers. The audience includes endocrinologists, internists,
cardiologists, neurologists, nephrologists, podiatrists, ophthalmologists,
family physicians, nurse practitioners, nurse educators, and physician
assistants.
Solomon Tesfaye
Christopher H. Gibbons
Rayaz Ahmed Malik  •  Aristidis Veves
Editors

Diabetic Neuropathy
Advances in Pathophysiology
and Clinical Management

Third Edition
Editors
Solomon Tesfaye Christopher H. Gibbons
Royal Hallamshire Hospital Beth Israel Deaconess Medical Center
Sheffield Teaching Hospitals Boston, MA, USA
and the University of Sheffield
Director of Diabetes Research Aristidis Veves
Sheffield, UK Department of Surgery
Beth Israel Deaconess Medical Center
Rayaz Ahmed Malik Boston, MA, USA
Weill Cornell Medicine
Cornell University
Doha, Qatar

ISSN 2197-7836     ISSN 2197-7844 (electronic)


Contemporary Diabetes
ISBN 978-3-031-15612-0    ISBN 978-3-031-15613-7 (eBook)
https://doi.org/10.1007/978-3-031-15613-7

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Switzerland AG 2023
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher,
whether the whole or part of the material is concerned, specifically the rights of translation,
reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any
other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, expressed or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This Humana imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
To Sophie, Aida and Sami—thanks for your unfailing support
Contents

Part I Clinical Features and Diagnosis

Introduction����������������������������������������������������������������������������������������������   3
Andrew J. M. Boulton

The Epidemiology of Diabetic Neuropathy ������������������������������������������   5
Christian Stevns Hansen, Laura L. Määttä, Signe Toft Andersen,
and Morten H. Charles

Clinical Features of Diabetes Neuropathies������������������������������������������  37
Gordon Sloan, Qi Pan, Ling Gao, Lixin Guo, and Solomon Tesfaye

Neuropathy in Type 1 and Type 2 Diabetes ������������������������������������������  51
Gulcin Akinci, Dustin Nowacek, and Brian Callaghan

Clinical Diagnosis of Diabetic Peripheral Neuropathy������������������������  67
Bruce A. Perkins and Vera Bril

Diagnostic Techniques for Diabetic Peripheral Neuropathy����������������  93
Long Davalos, Amro Stino, and A. Gordon Smith

Sensory Profiles and Diabetic Neuropathy�������������������������������������������� 113
Juliane Sachau, Manon Sendel, and Ralf Baron
Neurotrophic Factors in the Pathogenesis
and Treatment of Diabetic Neuropathy������������������������������������������������� 127
Nigel A. Calcutt

Treatment Induced Neuropathy of Diabetes ���������������������������������������� 157
Nadia McMillan and Christopher H. Gibbons
Asymmetric Diabetic Neuropathy: Radiculoplexus
Neuropathies, Mononeuropathies, and Cranial Neuropathies������������ 165
Pariwat Thaisetthawatkul and P. James B. Dyck

Motor Neuropathy in Diabetes �������������������������������������������������������������� 183
Karolina Snopek Khan and Henning Andersen
Cardiovascular Autonomic Neuropathy������������������������������������������������ 203
Lynn Ang and Rodica Pop-Busui

vii
viii Contents

Psychosocial Aspects of Diabetic Neuropathy:


From Description to Interventions �������������������������������������������������������� 221
Loretta Vileikyte and Frans Pouwer

Part II Pathophysiology


The Genomics of Diabetic Neuropathy�������������������������������������������������� 239
Abirami Veluchamy, Blair H. Smith, and David L. Bennett

Metabolic Mechanisms in Diabetic Neuropathy ���������������������������������� 253
Mark Yorek
Mechanisms of Nerve Injury in Diabetes:
Dyslipidemia, Bioenergetics, and Oxidative Damage �������������������������� 279
Stephanie A. Eid, Mohamed Noureldein, Masha G. Savelieff,
and Eva L. Feldman

Targeting the Mitochondrion in Diabetic Neuropathy ������������������������ 307
Ahmad Hedayat, Krish Chandrasekaran, Lindsay A. Zilliox,
and James W. Russell
Diabetic Sensory Neurons, Dorsal Root Ganglia,
and Neuropathy���������������������������������������������������������������������������������������� 327
Aparna Areti and Douglas W. Zochodne

Micro- and Macrovascular Disease in Diabetic Neuropathy �������������� 351
Lihong Chen and Aristidis Veves

The Spinal Cord in Diabetic Neuropathy���������������������������������������������� 363
Andrew G. Marshall, Anne Worthington, and Corinne G. Jolivalt

Brain Changes in Diabetes and Cognitive Dysfunction������������������������ 381
Geert Jan Biessels
Lifestyle and Dietary Modifications: Relevance
in the Management of Diabetic Neuropathy������������������������������������������ 397
Jonathan Enders and Douglas E. Wright

Pathophysiology of Neuropathic Pain���������������������������������������������������� 415
Andreas C. Themistocleous and Miroslav Misha Backonja

Central Nervous System Involvement in Painful
Diabetic Neuropathy�������������������������������������������������������������������������������� 427
Dinesh Selvarajah, Joyce Lim, Kevin Teh, Xin Chen,
Jing Wu, and Solomon Tesfaye

Part III Clinical Consequences and Treatments


Characteristics and Treatment of Painful Diabetic Neuropathy �������� 441
Sandra Sif Gylfadottir and Nanna Brix Finnerup
Orthostatic Hypotension and Sudomotor
Dysfunction in Diabetes�������������������������������������������������������������������������� 453
Lauren F. Fanty and Christopher H. Gibbons
Contents ix

Gastrointestinal Neuropathy������������������������������������������������������������������ 471


Karen L. Jones, Chinmay S. Marathe, Tongzhi Wu,
Christopher K. Rayner, and Michael Horowitz

Diabetic Neuropathy: Clinical Management—Genitourinary
Dysfunction in Diabetes�������������������������������������������������������������������������� 491
Vincenza Spallone, Enrico Finazzi Agrò, Roberta Centello,
Claudio Lecis, Luca Orecchia, and Andrea M. Isidori
Index���������������������������������������������������������������������������������������������������������� 531
Part I
Clinical Features and Diagnosis
Introduction

Andrew J. M. Boulton

1 Early History of the Diabetic acknowledge a direct link between diabetes and


Neuropathies any nervous system disorder.
It was not until 1864 that Marchal de Calvi
Whereas the first ever records of diabetes appear correctly described the relationship between dia-
to be found in ancient Egypt around 1550 BC, the betes and peripheral neuropathies [5]. Indeed, it
history of the diabetic neuropathies is relatively was the well-known Parisian physician Jean-­
recent. Indeed, the first references to possible Martin Charcot who acknowledged the impor-
involvement of the nervous system as a compli- tance of the observation by Marchal de Calvi:
cation of diabetes did not appear until the end of Charcot also described that in some diabetic
the eighteenth century [1]. It was Dr John Rollo, cases, there are signs of neurological disorders
who was a surgeon in the British Royal Artillery, [5]. It was towards the end of the nineteenth cen-
who wrote two texts on diabetes and in one of tury that Charcot summarised the clinical fea-
them described a patient with “burning pains in tures of diabetic peripheral neuropathy [6].
the palms and hands and the soles of the feet with Another pivotal observation at the end of the
weakness and complains of painful and gnawing nineteenth century was made by a surgeon in my
sensations in the leg” [2, 3]. However, as pointed hometown of Nottingham, UK.  It was Dr T
out by Ward [3, 4], reports that such patients with Davies Pryce that described what we now know
the symptoms as described above, “had a good as neuropathic foot ulcers and stated that these
night’s sleep” this would not be typical of painful were a consequence of peripheral nerve degen-
diabetic neuropathy. Moreover, Rollo did not eration [7].
One of the best descriptions of painful neuro-
pathic symptoms was made in 1894 by Pavy, a
physician at the Middlesex hospital in London
[8], who stated that they were of “of a burning and
A. J. M. Boulton (*) unremitting character”. Pavy also observed that
Division of Diabetes, Endocrinology and the pains were generally worse during the night.
Gastroenterology, University of Manchester,
Manchester, UK By the end of the nineteenth century, the neu-
ropathies of diabetes were well recognized:
Manchester Diabetes Centre, Manchester Royal
Infirmary, Manchester, UK indeed, it was stated that it was rare to meet a
case of diabetes without some evidence of ner-
University of Miami, Miami, FL, USA
vous disturbance [1].
International Diabetes Federation, Brussels, Belgium
e-mail: ABoulton@med.miami.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 3


S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_1
4 A. J. M. Boulton

2 The Twentieth Century In conclusion, most of our understanding of


the diabetic neuropathies has evolved over the
It was RT Williamson, a physician at the last 250 years. However, much progress has been
Manchester Royal Infirmary, who introduced made in the first 20 years of this century and
physiological measurements such as measure- much of this is covered by the excellent chapters
ment of vibration perception, into the assess- to be found in this up-to-date review of all aspects
ment of neuropathy early in the twentieth century of the diabetic neuropathies. I know each and
[9]. Coincidentally, I joined the same hospital as every one of the authors and can attest that they
a physician in 1986—and my group confirmed are all experts in their own fields and this will
that loss of vibration sensation was the most reli- provide a most useful volume to update knowl-
able predictor of foot ulceration in diabetes [10]. edge on the many various conditions that are
After the discovery of insulin, the understand- included under the title of diabetic neuropathies.
ing of the neuropathies increased, and a series of
important publications appeared from US cen-
tres. First, Jordan, in a series of patients, observed References
that neuropathic symptoms did not always match
signs in diabetic patients [11]. 1. Skljarevski V. Historical aspects of the diabetic neu-
ropathies. In: Veves A, Malik RA, editors. Diabetic
A decade later Wayne Rundles from the neuropathy: clinical management. 2nd ed. Totowa:
University of Michigan published observations Humana Press; 2001. p. 1–6.
on 125 cases of diabetic neuropathy, and not only 2. Rollo J. Cases of the diabetes mellitus with the results
proposed an association between glycaemic con- of the trials of certain acids. London: T.  Gillet for
C. Dilly; 1798.
trol and neuropathy, but also described the fea- 3. Ward JD. A history of diabetic neuropathy in the 19th
tures of autonomic dysfunction in diabetes [12]. century. In: Hotta N, Greene DA, Ward JD, et al., edi-
Again coincidentally, the same institution tors. Diabetic neuropathy: new concepts and insights.
remains active in research into the neuropathies Amsterdam: Elsevier; 1995. p. 89–95.
4. Ward JD.  Historical aspects of diabetic peripheral
with no fewer than three authors in the book com- neuropathy. In: Boulton AJM, editor. Diabetic neu-
ing from the University of Michigan. ropathy. Bridgewater: Aventis; 2001. p. 6–15.
It was Sven-Erik Fagerberg, from 5. De Calvi M. Recherches sur les accidents diabetiques.
Gothenburg, who in 1959 published extensive Paris: Asselin; 1864.
6. Charcot JM.  Sur un cas de paraplegie diabetique.
studies on 356 cases of diabetic neuropathy, Arch Neurol. 1890;19:305–35.
and in about half, he obtained and studied sec- 7. Pryce TD.  Perforating ulcers of both feet associ-
tions of peripheral nerve, observing substantial ated with diabetes and ataxic symptoms. Lancet.
abnormalities of the nerve microvasculature 1887;2:11–2.
8. Pavy FW. The Croonian lecture on new departures in
[13]. He proposed the potential of a common diabetes. Br Med J. 1894;1:1349–50.
aetiopathogenesis of neuropathy, retinopathy, 9. Williamson RT.  A clinical lecture on the vibrating
and nephropathy. Sadly, the closing years of the sensation in diseases of the nervous system: deliv-
twentieth century may well be remembered for ered at the Manchester Royal Infirmary. Br Med J.
1907;2:125–7.
numerous clinical trials of putative new thera- 10. Young MJ, Breddy JL, Veves A, Boulton AJM.  The
pies that aimed to tackle the underlying patho- prediction of diabetic neuropathic foot ulceration
genesis of neuropathy, none of which showed using vibration perception thresholds: a prospective
clinical efficacy. study. Diabetes Care. 1994;17:557–60.
11. Jordan WR. Neuritic manifestations in diabetes mel-
To end this historical review on a more posi- litus. Arch Intern Med. 1936;57:307.
tive note, the art of clinical medicine remains 12. Rundles RW.  Diabetic neuropathy: general review
important in the diagnosis of neuropathy, and it with report of 125 cases. Medicine. 1945;24:111–60.
was my mentor and renowned neuropathy 13. Fagerberg S-E.  Diabetic neuropathy: a clinical and
histological study on the significance of vascular
researcher John D Ward (1935–2019), who affections. Acta Med Scand. 1959;164(345):1–97.
described a clinical sign of autonomic neuropa- 14. Ward JD, Boulton AJM, Simms JM, et al. Venous dis-
thy in the diabetic foot—distended dorsal foot tension in the diabetic foot – physical sign of arterio-
veins [14]. venous shunting. J R Soc Med. 1983;76:1011–4.
The Epidemiology of Diabetic
Neuropathy

Christian Stevns Hansen, Laura L. Määttä,
Signe Toft Andersen, and Morten H. Charles

1 Introduction independently associated with cardiovascular


morbidity and mortality [1].
Diabetic neuropathy is a common and severe While the incidence of major diabetic compli-
complication of diabetes. It is associated with cations, such as cardiovascular disease, renal dis-
substantial socioeconomic and personal conse- ease, blindness and amputation, has been
quences. The complication encompasses a het- decreasing steadily in most countries for the last
erogeneous group of neuropathies, which affect few decades [2], we do not know if this is true for
various parts of the nervous system, leading to a diabetic neuropathy. Despite its severity, the epi-
wide range of different symptoms and signs of demiological data at hand is not as substantial as
nerve fibre dysfunction. In addition, different for many other diabetic complications. This is
types of neuropathy are independent risk factors linked to a lack of international consensus on the
for various end-organ manifestations. For exam- diagnostic criteria for this heterogeneous compli-
ple, distal symmetrical polyneuropathy (DSPN) cation and to the lack of robust, long-term
is an independent risk factor for peripheral arte- population-­based studies reporting the incidence
rial disease and peripheral amputations, while of different neuropathies.
cardiovascular autonomic neuropathy (CAN) is Our epidemiological understanding of dia-
betic neuropathy is mainly based on cross-­
sectional studies performed at different time
C. S. Hansen
Steno Diabetes Center Copenhagen, Herlev, Denmark points. This allows for point estimates of preva-
lence. In addition, data from large longitudinal
L. L. Määttä
Danish Pain Research Center, Department of Clinical cohort studies and randomised trials have shed
Medicine, Aarhus University, Aarhus, Denmark light on the natural history of diabetic neuropa-
Steno Diabetes Center Aarhus, Aarhus University thy. However, data on indices rate ratios of dia-
Hospital, Aarhus, Denmark betic neuropathy over time are non-existent.
S. T. Andersen Thus, it cannot be documented if incidence rates
Steno Diabetes Center Aarhus, Regional Hospital are improving.
Gødstrup, Herning, Denmark Diabetic neuropathy can be classified into four
M. H. Charles (*) groups: symmetric polyneuropathy, autonomic
Steno Diabetes Center Aarhus, Aarhus University neuropathy, polyradiculopathies, and mononeu-
Hospital, Aarhus, Denmark
ropathies. In this chapter, we will present an in-­
Department of Public Health, Aarhus University, depth analyses of the epidemiology of the two
Aarhus, Denmark
most prevalent forms of diabetic neuropathy: (1)
e-mail: mc@ph.au.dk

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 5


S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_2
6 C. S. Hansen et al.

Distal symmetrical polyneuropathy (DSPN) and ropathy in both type 1 (T1D) and type 2 diabetes
(2) cardiovascular autonomic neuropathy (CAN). (T2D) constituting approximately 80–90% of
We also provide a short overview of other forms diabetic neuropathies [1]. Diabetic DSPN is
of diabetic neuropathy. termed typical diabetic neuropathy while the
less common forms are called atypical diabetic
neuropathies [3]. In fact, DSPN is the most
1.1 Strategy for Reviewing common form of neuropathy, irrespective of
Relevant Literature aetiology [4], and diabetes is the most common
cause of DSPN [4].
This chapter is a review of major studies carried Common symptoms of DSPN include neuro-
out in the past four decades. In addition to report- pathic pain, numbness, tingling, prickling, tem-
ing data from landmark studies such as UKPDS, perature insensitivity, and weakness [1, 3].
ACCORD, EURODIAB, ADDITION-Denmark, Symptoms normally start in the toes and feet and
the Search Study, DCCT/EDIC, MONICA/ spread proximally, involving the fingers and
KORA, and others, we also performed PubMed hands in more severe cases. This typical symmet-
searches relating to DSPN and CAN.  However, ric distribution of length-dependent neuropathic
the chapter does not have an exhaustive list of symptoms is described as a stocking-and-glove
studies in the field of diabetic neuropathy research. pattern [1, 3]. Dysesthesias and pain are com-
In addition to a presentation of the prevalence and monly the first symptoms and as DSPN pro-
natural history of diabetic neuropathy we aim to gresses, motor involvement may appear [1, 3, 5].
elucidate risk factors for the complication. DSPN has a major impact on the life and health
For DSPN, relevant literature was obtained of affected people due to invalidating pain, com-
from recent comprehensive reviews on diabetic plicating foot ulcers, falls, and amputations lead-
neuropathy and on the epidemiology of diabetic ing to a decreased quality of life [1, 6, 7].
neuropathy. We performed additional searches on No single decisive definition of DSPN exists.
PubMed using the mesh terms “diabetic neuropa- The American Diabetes Association’s (ADA)
thies”, “prevalence”, “incidence”, and “epidemi- definition of DSPN for clinical practice is “the
ology” from 2013 and onwards. From the presence of symptoms and/or signs of peripheral
prevalence-related search, studies with over 400 nerve dysfunction in people with diabetes after
participants were included. the exclusion of other causes” [1]. ADA recom-
We performed PubMed CAN searches using mends that the assessment for DSPN should
the key words “diabetes”, “autonomic neuropa- include a careful history and assessment of both
thy”, “incidence”, and “prevalence”. Studies per- small (temperature or pinprick sensation) and
formed prior to 1980 were not assessed and we large fibre function (vibration perception and
did not include cohorts with less than 80 partici- 10-g monofilament) [8]. Further evaluation by
pants. However, smaller cohorts from non-­ nerve conduction studies (NCS) or skin biopsies
western populations were included due to a to assess intraepidermal nerve fibre density
paucity of studies reported from these regions. (IENFD) is rarely needed and is primarily
reserved for research purposes. The Toronto
Consensus Panel criteria for DSPN are widely
2 Diabetic Distal Symmetric accepted as the current gold standard for diag-
Polyneuropathy nosing DSPN in a research context [3]. The
Toronto criteria define hierarchical categories of
It is estimated that half of all people with diabe- DSPN graded as possible (symptoms or signs),
tes develop neuropathy during their disease [1]. probable (≥2 following: symptoms, signs, abnor-
Half of these cases may be asymptomatic [1]. mal ankle reflexes), or definite (symptoms and/or
Distal symmetric sensorimotor polyneuropathy signs together with a confirmatory test of DSPN
(DSPN) is by far the most common type of neu- (usually NCS, IENFD)) [3, 9].
The Epidemiology of Diabetic Neuropathy 7

There is emerging evidence that DSPN can be estimating the prevalence of DSPN is popula-
present even prior to the diagnosis of diabetes tion-based survey [17].
with a prevalence of 10–30% in people with pre- In general, age and diabetes duration are
diabetes or metabolic syndrome [1, 10–17]. important factors to consider when comparing
However, such cases of DSPN might not be diag- the prevalence of DSPN in different studies, irre-
nosed unless small fibre impairment is examined: spective of their setting.
the natural history of DSPN is hypothesised to
start as small fibre damage and proceed towards
large fibre damage [1, 5, 18, 19]. 2.2 Prevalence of DSPN

The prevalence of neuropathy in the general pop-


2.1 Issues to Consider When ulation is estimated to be 1–8% and approxi-
Evaluating Epidemiological mately half of these cases are due to diabetes [4,
Studies of DSPN 6, 22, 23]. The prevalence of DSPN in T1D and
T2D for selected studies and settings is given in
Summarising the prevalence and incidence of Table  1. These studies reveal a varying preva-
DSPN based on existing studies is not straight lence of DSPN, from 1.5% [30] to 75.1% [67].
forward for several reasons [17]. First, the vary- Very similar ranges exist when comparing T1D
ing pattern of nerve fibre damage can give rise to and T2D (T1D primary care/community-based:
a heterogeneous presentation of DSPN.  For 7–62.5% (median 20.8%) and hospital based:
example, one patient may have decreased vibra- 2.6–55.1% (median 22.1%); and T2D primary
tion perception threshold (VPT) due to large fibre care/community-based: 1.5–63% (median
damage, while another patient may have intact 27.4%) and hospital based: 18.1–75.1% (median
VPT but decreased pinprick or temperature sen- 34.4%). The median values are calculated from
sation due to small fibre dysfunction. Hence, the the studies included in Table 1 with large varia-
chosen method for testing DSPN has an impact tion in age, diabetes duration, diagnostic tests,
on prevalence estimates [17, 20]. An example etc.). Furthermore, it is worth noting that studies
illustrating this issue is the baseline examination defining DSPN by NCS tend to report a higher
of young adults with T1D in the Diabetes Control prevalence of DSPN compared to other diagnos-
and Complications Trial (DCCT). The prevalence tic tests, underpinning the high sensitivity of
of DSPN varied from 0.3% (abnormal reflexes, NCS to detect DSPN [29, 31, 44]. Overall, the
sensory signs, and neuropathic symptoms) to prevalence of DSPN increases with diabetes
21.8% (abnormal NCS in at least two nerves) in duration and with increasing age despite the set-
the standard-care group depending on which test ting or testing method.
was used to define DSPN [17, 21]. In recognition In newly diagnosed diabetes, the prevalence
of this issue, the current clinical guidelines for of DSPN tends to be lower [25, 31–34, 41, 45–
diagnosing DSPN recommend a combination of 47, 65, 68]. A population-based survey of DSPN
tests and symptoms to detect this complication. in Mauritius reported a prevalence as low as 3.6%
Second, several available tests for evaluat- in newly diagnosed T2D (N = 414) where DSPN
ing DSPN are subjective in nature, requiring was defined using age-specific VPT [34].
people to interpret an external stimulus (vibra- Population-based samples from Egypt and India
tion, hot or cold application, etc.) and can thus show a prevalence of 14% (N = 125) and 19.5%
be hard to reproduce [17]. Third, the preva- (N = 338), respectively, in people with newly
lence varies between different study-popula- diagnosed T2D. These studies also used VPT to
tions. Hospital-­ based study cohorts can be detect DPSN but did not apply age-specific refer-
expected to have a higher prevalence of DSPN ence values [33, 41]. The ADDITION-Denmark
compared with primary-­ care or community- study, which included individuals with screen-­
based cohorts [17]. The optimal method for detected T2D (N = 1445), reported a prevalence
8
Table 1  Prevalence of DSPN in type 1 and 2 diabetes
Participants DM duration Mean age
Study site (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 1 (P) USA 363 ≥2 of the following: 34 19.9 28.4 DM duration, HbA1c, smoking status,
EDC [24] symptoms, signs (light HDL-c
touch, pinprick, VPT,
proprioception, muscle
weakness, gait), knee/
ankle reflexes
Type 1 (P) Italy [25] 379 Symptoms and ≥1 sign 28.5 NA NA (15–59 NA
OR ≥2 signs year)
(VPT, ankle/knee
reflexes, foot ulcers, DB,
postural hypotension)
Type 1 (P) Denmark [26] 339 VPT 62.5 13 20 Male sex, age, increased albumin
excretion rate
Type 1 (P) USA [27] 5936 MNISq ≥ 4 11 18 (median) 39 Age, HbA1c, DM duration, lower
socioeconomic status, CVD, smoking,
hypertriglyceridemia, BMI,
retinopathy, reduced eGFR, Charcot
neuroarthropathy, recent severe
hypoglycaemia and/or DKA
Type 1 (P) Scotland [28] 5558 MNNIq ≥ 4 13 20.5 44.7 Age, DM duration, waist-to-hip ratio,
lipids, HbA1c, smoking, low eGFR,
low HDL-c, socioeconomic status, ≥1
other DM complication (nephropathy,
retinopathy, CVD, recent severe
hypoglycaemia or DKA)
Type 2 (P) USA [29] 78 NCS 46.2 NA 62.1 NA
Type 2 (P) Finland [30] 132 (1) Symptoms (1) 1.5 0 Men 54.9 NA
(2) Knee/ankle reflexes (2) 2.3 Women
and ≥1 sign (light touch, (3) 15.2 57.1
pinprick, VPT) OR ≥2
signs
(3) NCS
C. S. Hansen et al.
Participants DM duration Mean age
Study site (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 2 (P) Finland [31] 132 NCS with or without Baseline: 8.3 10 56 Fasting Glc levels, lower insulin levels
symptoms 10-year at fasting and after oral Glc
follow-up: administration
41.9
Type 2 (P) USA 279 ≥2 of following: 25.8 NA NA (20–74 Age, DM duration, male sex, HbA1c
San Luis Valley Symptoms, ankle year)
Diabetes Study [32] reflexes, temp. sensation
Type 2 (P) Egypt [33] 384 VPT 21.9 NA NA (over HbA1c, age, female sex,
20 year) hypercholesterolemia
Type 2 (P) Mauritius [34] 433 VPT 12.7 6.6 55 DM duration, treatment with insulin or
oral hypoglycaemic agents, height,
fasting Glc, lower 2-h plasma insulin
Type 2 (P) Netherlands [35] 137 (1) Symptoms (1) 18 7.5 68 NA
(2) VPT (2) 53
The Epidemiology of Diabetic Neuropathy

(3) Knee/ankle reflexes (3) 62


(4) Temp. sensation (4) 63
Type 2 (P) Italy [36] 347 ≥2 symptoms and ≥2 of 19 NA NA (≥55 DM duration, fasting and post-prandial
following: “sensation, year) Glc
strength, tendon reflexes”
Type 2 (P) UK [37] 811 NDS ≥ 6 41.6 7.4 65.4 NA
Type 2 (P) Portugal [38] 93 Knee/ankle reflexes and 32.2 10.1 65.4 Age, DM duration, foot skin changes,
≥1 sign (VPT, MI/ischemia
monofilament, pinprick,
temp. sensation,
proprioception)
Type 2 (P) Malaysia [39] 134 NSS and NDS 50.7 8.5 56.5 Age, DM duration
Type 2 (P) Bahrain [40] 1477 ≥2 of following: 36.6 9.5 57.3 Age, DM duration, HbA1c, total
symptom score, sign cholesterol, smoking, BMI, waist
score (“pain”, “touch”, circumference, triglycerides,
temp. sensation, VPT), hypertension
VPT
Type 2 (P) India [41] 1291 VPT 27.8 5.9 +DSPN 60 Age, HbA1c, DM duration
–DSPN 47
(continued)
9
10
Table 1 (continued)
Participants DM duration Mean age
Study site (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 2 Germany 195 MNSI exam > 2 28 NA 66.8 Age, waist circumference, PAD
predominantly MONICA/KORA
(P) [42]
Type 2 (P) Sweden [43] 156 VPT and/or 34 7.0 61.7 PAD, age, male sex, lower
monofilament HDL-cholesterol
Type 2 (P) Taiwan [44] 133 NCS 29.3 +DSPN 3 +DSPN Age, renal insufficiency, HbA1c,
–DSPN 2 66.2 fasting Glc, systolic BP
–DSPN
54.9
Type 2 (P) UK 3867 (1) Ankle reflexes Baseline: 0 Baseline NA
UKPDS [45] (2) VPT (1) INT 11.8; 53.3
CON 11.4
12-year
follow-up:
(1) INT 35;
CON 37
(2) INT 30.2;
CON 32.8
Type 2 (P) Denmark 1445 MNSIq ≥ 4 13.1 0 NA (–DPN NA
ADDITION-DK 60.8)
Baseline [46]
Type 2 (P) Denmark 1161 (1) Monofilament and/or (1) INT 30.1; 6 INT 59.6, NA
ADDITION-DK 6-yr VPT CON 34.8 CON 59.9
follow-up [47] (2) MNSIq ≥ 7 (2) INT 8.7;
CON 9.3
Type 2 (P) Denmark 452 NCSa and ≥ 1 symptom 27.0 +DSPN 12.9 70.9 Baseline HbA1c, steeper increases in
ADDITION-DK and/or sign (ankle reflex, –DSPN 12.7 HbA1c over time, weight, waist
13-yr follow-up [48] VPT, monofilament) circumference, BMI
Type 2 (P) Brazil [49] 551 (1) NSS = 5–6 OR NSS (1) 6.3 10 65 DM duration
= 3–4 and NDS = 6–8 (2) 14.3
(2) Monofilament
Type 1 + 2 (P) Australia [50] T1 179 Pinprick T1 8.4 T1 12.6 T1 33.8 DM duration, age at DM diagnosis
T2 904 T2 16.8 T2 7.2 T2 63.6
C. S. Hansen et al.
Participants DM duration Mean age
Study site (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 1 + 2 (P) USA [51] T1 102 ≥2 of following: T1 54 T1 14.5 T1 41 NA
T2 278 symptoms (NSS [52]), T2 45 T2 8.1 T2 63.2
signs (NDS [52]), NCS, (median)
VPT, CDT, DB, VAL,
with ≥1 being NCS, DB
or VAL
Type 1 + 2 USA [53] T1 124 Symptoms T1 30.2 T2 T2 DM duration, hypertension,
(P) T2 2268 T2 37.9 +DSPN 12.0 +DSPN hyperglycaemia, glycosuria
–DSPN 9.6 61.8
–DSPN
61.7
Type 1 + 2 UK [54] T1 213 ≥2 of the following: T1 12.7 NA NA T1: height, retinopathy
(P) T2 864 symptoms, light touch, T2 17.2 T2: age, height, alcohol, HbA1c,
pinprick, ankle reflexes, retinopathy
The Epidemiology of Diabetic Neuropathy

VPT (only for <70 year


olds)
Type 1 + 2 USA T1 1734 MNSI exam >2 T1 7 T1 7.2 T1 18 T1: Age, DM duration, poor glycaemic
(P) SEARCH [55] T2 258 T2 22 T2 7.9 T2 22 control, smoking, diastolic BP, obesity,
increased LDL-c and triglycerides,
lower HDL-c
T2: Age, male sex, DM duration,
smoking, lower HDL-c
Type 1 (H) Europe 3250 ≥2 of the following: 28.5 14.7 32.7 Age, DM duration, HbA1c, height,
EURODIAB Baseline symptoms, ankle/knee background or proliferative diabetic
[56] reflexes, VPT, HRV and/ retinopathy, cigarette smoking,
or postural hypotension reduced HDL-c, CVD, diastolic BP,
severe ketoacidosis, raised fasting
triglyceride, microalbuminuria
(continued)
11
12
Table 1 (continued)
Participants DM duration Mean age
Study site (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 1 (H) Europe 1172 Same as above Cumulative – – HbA1c, change in HbA1c during
EURODIAB incidence follow-up, DM duration, elevated total
7.3-year follow-up 23.5 and LDL-c, elevated triglycerides,
[57] higher BMI, higher von Willebrand
factor, elevated urinary albumin
excretion rate, hypertension, smoking,
CVD at baseline
Type 1 (H) USA and Canada 1186b NCS and ≥2 of the Baseline: Baseline: INT Baseline: Higher mean HbA1c, age, DM
DCCT/EDIC following: symptoms, INT 7, 6; CONV 6 INT 27; duration, height, macroalbuminuria,
(Summarising signs (light touch, CONV 5 Closeout: INT CONV 27 higher mean pulse rate, β-blocker use,
DPN-findings: Albers proprioception, temp. Closeout: 12; CONV 12 Closeout: sustained albuminuria
et al. [58], Martin sensation, pinprick), INT 9; EDIC year INT 34;
et al. [59], risk factors ankle reflexes CONV 17 13/4: INT 26; CONV 33
[60]) EDIC year CONV 26 EDIC year
13/4: INT 25; 13/4: INT
CONV 35 48; CONV
47
Type 1 (H) Denmark [61] 156 (1) Confirmed: symptoms (1) 2.6 11.3 22 None
or signs (monofilament, (2) 55.1
VPT, pinprick) and NCSa
or ECSc
(2) Subclinical: abnormal
NCS or ECS
Type 2 (H) USA [62] 775 Monofilament 50 11 62 Height, previous foot ulcer, age,
HbA1c, alcohol, smoking, albumin
level adjusted for serum creatinine
Type 2 (H) Tanzania [63] 153 Score based: 28 5.2 44.2 NA
Several signs (VPT,
“sensibility”, hallux
flexion, hair loss, foot
deformities, dry/cracked
skin) OR signs and
symptoms (incl.
claudication)
Type 2 (H) Saudi-Arabia [64] 375 Pinprick and/or vibration 20 8 (median) 50 (median) NA
sensation
C. S. Hansen et al.
Participants DM duration Mean age
Study site (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 2 (H) Netherlands [65] 73 VPT 50 6.6 (median) 65.9 NA
Type 2 (H) France [66] 427 Ankle reflexes and/or 31.6 10.6 56.9 NA
VPT
Type 2 (H) Sweden [64] 79 Pinprick and/or vibration 19 11.5 (median) 58 (median) NA
sensation
Type 2 (H) Iran [67] 810 ≥1 of following: 75.1 8.2 52.7 Age, proteinuria, DM duration,
pinprick, VPT, light insulin-treatment, retinopathy, IHD
touch, proprioception,
ankle reflexes, muscular
power
Type 2 (H) China [68] 1067 VPT and/or 18.3 7.2 59.8 Retinopathy, HbA1c, DM duration,
monofilament age
Type 2 (H) Denmark 160 VPT Baseline 34.4 INT 5.5 55.1 NA
Steno 2 [69] CON 6.0 (40–65) (INT (targeting hyperglycaemia, risk
The Epidemiology of Diabetic Neuropathy

factors and behaviour) without


significant effect on DSPN)
Type 2 (H) USA 10,251 MNSI exam >2 Baseline: 10 62 INT glycaemic therapy decreased
ACCORD [70] INT 43; DSPN risk
CON 42
5-year
follow-up:
INT 55.6;
CON 58.6
Type 2 (H) China [71] 562 NCS and symptoms and/ 18.1 5.6 56.4 Age, DM duration, hypertension,
or signs (ankle reflex, insulin resistance index, HbA1c,
VPT, monofilament, HbA1c variability, u-ACR
temp. sensation, pinprick,
proprioception
Type 1 + 2 Italy [72] 8757 ≥2 points: VPT, ankle 32.3 12.1 (data only 58.8 NA
(H) reflexes, foot ulcer, other from records of
foot deformity/dry skin/ 1227
infection (1 point per participants)
abnormality per side
(L/R))
(continued)
13
14
Table 1 (continued)
Participants DM duration Mean age
Study site (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 1 + 2 (H) Germany, Austria, T1 647 ≥2 of following: T1 17.1 T1 10.3 T1 33 T1 + T2: Age, retinopathy,
Switzerland [73] T2 524 symptoms, knee/ankle T2 34.8 T2 9.0 T2 54 macroangiopathy, CAN
reflexes, VPT (medians) (medians) T1: albuminuria, HbA1c
Type 1 + 2 (H) UK [74] T1 2414 NDS ≥ 6 OR NDS = 3–5 T1 22.7 T1 13 T1 45 Age, DM duration
T2 3949 and NSS ≥ 5 T2 32.1 T2 6 T2 63
(medians) (medians)
Type 1 + 2 Iran [75] T1 79 NDS ≥ 6 OR NDS = 3–5 T1 21.5 T1 9.5 T1 28.2 T1: DM duration, education level
(H) T2 521 and NSS ≥ 5 T2 49.3 T2 9.2 T2 57.0 T:2 history of foot ulcer, age, DM
duration, weight, education level, male
sex
Type 1 + 2 Spain [76] T1 348 NDS ≥ 6 OR NDS = 3–5 T1 12.9 T1 13.8 T1 30.5 T1: DM duration
(H + P) T2 2296 and NSS ≥ 5 T2 24.1 T2 9.7 T2 61.3 T2: age, DM duration
BMI body mass index, BP blood pressure, CDT cooling detection threshold, CON conventional treatment, CVD cardiovascular disease, DB deep breathing, DKA diabetic ketoaci-
dosis, DM duration diabetes mellitus duration, DN4 Douleur Neuropathique 4 Questions, ECS electrochemical skin conductance, Glc glucose, H Hospital/out-patient based,
HbA1c hemoglobin A1c, HDL/LDL/total-c high density lipoprotein/low density lipoprotein/total chlolesterol, IHD ischemic heart disease, INT intensive treatment, MI myocar-
dial infarction, MNSI examination Michigan neuropathy screening instrument examination, MNSIq Michigan neuropathy screening instrument questionnaire, NCS nerve conduc-
tion studies, NNS and NDS Neuropathy symptom score and Neuropathy disability score (Young et al. [74]), P primary care/community-based, PAD peripheral arterial disease,
S-LANSS self-report Leeds Assessment of Neuropathic Symptoms and Signs (S-LANSS) pain scale, u-ACR urin albumin-creatinine ratio, VAL Valsalva’s manoeuvre, VPT
vibration perception threshold
a
NCS measured with handheld DPNCheck©-device
b
Number of participants at EDIC year 13/14, mean age and DM duration reported for these
c
ECS measured with Sudoscan©-device
C. S. Hansen et al.
The Epidemiology of Diabetic Neuropathy 15

of 13.1% at the time of diabetes diagnosis using (EDIC), used a robust definition of DSPN requir-
the Michigan Neuropathy Screening Instrument ing ≥2 of following in addition to abnormal NCS:
questionnaire (MNSIq) to define DSPN [46]. symptoms, peripheral sensory signs, and abnor-
Similarly, the prevalence of DSPN in the United mal ankle reflexes. The prevalence in the conven-
Kingdom Prospective Diabetes Study (UKPDS) tional treatment group was 5% at baseline,
of 3,867 people with newly diagnosed T2D was increasing to 35% in the EDIC year 13/14 [58,
11.5%, defined using VPT [45]. Out-patient 59].
clinic-based samples show prevalence’s ranging In T2D, studies conducted in primary care or a
from 6.4% [68] to 39% [65] in people with newly community-based setting predominantly show a
diagnosed T2D.  In summary, estimates of the DSPN prevalence of between 20% and 50%,
prevalence of DSPN in newly diagnosed T2D again varying by diabetes duration and the
range from 4% and 40%, with lower prevalence method used for detecting DSPN.  A purely
(<20%) in larger study-populations. symptom-based study of more than 2200 people
It is clear from the summary data presented in with T2D in the USA, with a T2D duration of 10
Table 1 that the comparison of DSPN prevalence years, reported a DSPN prevalence of 37.9%
between studies is challenging. When evaluating [53]. In contrast, symptom-based DSPN preva-
studies of T1D in primary care or community-­ lence early in the course of T2D is as low as 1.5%
based cohorts, a DSPN prevalence of 7% was [30]. The UKPDS (newly diagnosed T2D) and
found in young people with T1D in the large ADDITION-Denmark study (screen-detected
population-based cohort (N = 1734) of the T2D) also describe relatively low DSPN preva-
SEARCH for Diabetes in Youth-study. Here, lences of 11.5% and 13.1%, respectively [45, 46].
DSPN was assessed using the MNSI examination In contrast, the Rochester Diabetic Neuropathy
[55]. In contrast, a smaller Danish study (N = Study cohort, with its robust DSPN definition,
339) reported a prevalence of 62.5% in young found a prevalence of 45% in people with T2D
people with T1D based on VPT [26]. Recently, with a diabetes duration of approximately 8 years
DSPN prevalence was estimated using the [51]. An even higher prevalence of DSPN was
MNSIq in two large cohorts of nearly 6000 adults seen in a smaller primary care-based study from
with T1D in the USA [27] and Scotland [28] the Netherlands, which defined DSPN as abnor-
reporting very similar prevalences of 11% and mal temperature sensation with a prevalence of
13%, respectively. However, studies that define 63% (N = 137) in people with T2D [35]. By con-
DSPN using symptoms alone might underesti- trast, a larger sample (N = 904) evaluating DSPN
mate the true prevalence of DSPN by overlook- with pinprick in people with T2D in an Australian
ing the large fraction of asymptomatic cases. rural area found a prevalence of only 17%,
Finally, the population-based Rochester Diabetic although age and diabetes duration were similar
Neuropathy Study cohort found a DSPN preva- to the cohort in the Netherlands [50]. Although
lence of 54% in 102 people with T1D when using the mean age in most T2D studies is around 60
a more complex DSPN definition similar to the years, the SEARCH-study, which included 258
“definite DSPN” category of the Toronto classifi- young adults with T2D (mean age 22 years),
cation [51]. found a DSPN prevalence of 22%—a prevalence
Among hospital-based studies of cohorts with that is similar to that observed in older T2D pop-
T1D, the European IDDM Prospective ulations [55].
Complications Study (EURODIAB) of over 3000 The prevalence of DSPN in hospital-based
people showed a prevalence of 28.5% when studies of T2D are generally higher than the prev-
defining DSPN using symptoms and signs [56]. alence of DSPN in T1D or in primary care/
Another large multicentre study from the UK community-­ based samples of T2D.  Two large
found a prevalence of 22.7% in a cohort with Asian out-patient surveys report a DSPN preva-
T1D with a similar DSPN definition [74]. The lence of approximately 20% when evaluated
DCCT, and its observational follow-up study using VPT and/or monofilament [68, 77]. The
16 C. S. Hansen et al.

prevalence in an age- and T2D duration compa- reflects the area of damaged nerve fibres. Similar
rable multicentre study from Korea was higher to DSPN, the distribution of pain is most often
(33.5%). This is likely explained by the fact that found in a stocking-and-glove pattern. For this
both symptoms and signs were included their reason painful diabetic neuropathy will be
DSPN definition [78]. Similarly, an earlier referred to as painful DSPN (P-DSPN). Alike
UK-based multicentre study of nearly 4000 peo- DSPN, neuropathic pain can also be classified as
ple with T2D showed a prevalence of 32.1% possible, probable, and definite according to a
when using both symptoms and signs, or several hierarchical grading system proposed by the
abnormal signs [74]. The Action to Control Neuropathic Pain Special Interest Group of the
Cardiovascular Risk in Diabetes (ACCORD) trial International Association for the Study of Pain
of more than 10,000 people defined DSPN using (NeuPSIG) [80].
the MNSI examination and found a baseline In practice, the diagnosis of P-DSPN relies on
prevalence of 42%. This increased to 58.6% in the patient’s subjective description of pain. The
the conventional therapy group after 5 years [70]. pain is typically described as burning, prickling,
The high prevalence of DSPN in this cohort is lancinating, tingling or shooting and may occur
likely explained by the high cardiovascular risk together with paresthesias, hyperalgesia, and
profile of the participants. An exceptionally high allodynia and is often worse at night [1, 3, 81].
prevalence was reported in an Iranian study Pain may be the first symptom of DSPN that
including 810 people that defined DSPN using a alerts the patient to seek health care and it has a
clinical exam score where any single impairment, major impact on the quality of life of affected
including small fibre impairment, qualified for a people by interfering with daily activities, sleep,
DSPN diagnosis. The reported prevalence was and enjoyment of life [1, 7, 17, 81–83]. Despite
75.1% in this middle-aged population with a T2D this, P-DSPN might be underdiagnosed and
duration of 8 years [67]. undertreated as recent reports have shown that up
To conclude, DSPN is a highly prevalent com- to 80% of people with P-DSPN may be undiag-
plication of both T1D and T2D. There is a wide nosed [84, 85], stressing the importance of accu-
range of reported prevalences, with median prev- rate screening of DSPN and P-DSPN to
alences around 20–30% for both T1D and adequately manage the pain and its adverse
T2D. An increasing prevalence of DSPN is seen effects on quality of life [1].
with increasing diabetes duration in both T1D As for DSPN, the prevalence of P-DSPN var-
and T2D. In summary, varying testing methodol- ies widely but it is lower than the overall preva-
ogies and differences in population characteris- lence of DSPN. Selected studies (Table 2) report
tics prevent a simple comparison between DSPN a prevalence between 5.4% and 42.2% for studies
prevalences. However, it is estimated that an of T1D and T2D in both hospital- and primary-­
equal proportion of people with T1D and T2D care/community-based settings with a median
are affected by DSPN during the course of their prevalence of around 17%. A large community-­
disease [6, 17]. based UK cohort with a predominance of people
with T2D found a P-DSPN prevalence of 13.4%
in T1D and 21.5% in T2D [91]. Interestingly,
2.3 Painful Diabetic Neuropathy 26% of the non-DSPN group presented with
P-DSPN suspicious pain, which might indicate
Neuropathic pain is defined as “pain caused by a an early stage of nerve fibre damage that is not
lesion or disease of the somatosensory nervous detectable by clinical examination. Smaller pri-
system” [79]. In the case of painful diabetic neu- mary care/community-based studies from the UK
ropathy, the damage to the peripheral nervous and Germany that define P-DSPN with both clin-
system is due to diabetes. As for DSPN, no single ical signs and symptoms of pain, report P-DSPN
gold standard exists for the diagnosis of painful prevalences between 13.3% and 26.4% in T2D
diabetic neuropathy, where the pain location [82, 86, 87, 90].
Table 2  Prevalence of painful DSPN, and DSPN when stated, in type 1 and 2 diabetes
DM duration Mean age
Study site Participants (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 2, UK [82] 269 Toronto clinical scoring system >5 and 26.4 8 66.7 NA
P-DSPN (P) history suggesting neuropathic pain
Type 2, Germany [86] 195 MNSI exam >2 and neuropathic pain 13.3 NA 66.8 Age, weight, PAD
P-DSPN (P) (MNSIq)
Type 2, Germany [87] 214 MNSI exam >2 and neuropathic pain 21 NA 69 Waist circumference,
P-DSPN (P) (MNSIq) physical activity, PAD
Type 2, Denmark 5514 (1) Possible DSPN: MNSIq ≥ 4 (1) 18 4.6 64.1 (1) Female sex, age, DM
P-DSPN (P) DD2 [88] (2) Possible P-DSPN: bilateral pain in (2) 10 duration, BMI, smoking
the feet and DN4 questionnaire ≥3 (2) Smoking
Type 2, Denmark 5514 (1) Possible DSPN: ≥1 of following: (1) 62.2 . . NA
P-DSPN (P) DD2 [89] Estimates (2) 43.9
The Epidemiology of Diabetic Neuropathy

symptoms, signs (light touch, pinprick,


based sample temp. sensation, VPT), ankle reflexes (3) 22.7
of N = 389 (2) Probable DSPN: ≥2 of the above 3 (4) 5.4
(3) Definite DSPN: NCS or IENFD
and ≥1 of the above 3
(3) Definite P-DSPN: NCS or IENFD
and bilateral pain in feet, signs
Type 2, Korea [78] 3999 DSPN: MNSIq ≥ 3 and monofilament DSPN 33.5 P-DSPN 13.1 P-DSPN P-DSPN: age, female
P-DSPN (H) OR typical DSPN signs and symptoms of which 43.1 Non-P-­DSPN 63.6 gender, fasting Glc,
evaluated by study physician P-DSPN 11.4 Non-P-­ hypertension, previous
P-DSPN: DSPN and VAS ≥ 4 last 48 h DSPN 62.5 cerebrovascular events
or use of pain medication for pain in
legs and feet or legs and hands
Type 2, Taiwan [77] 2837 DSPN: Monofilament and/or VPT DSPN 21.3 10.1 63.9 DSPN: age, insulin
P-DSPN (H) P-DSPN: DSPN and DN4 ≥ 4 of which treatment,
P-DSPN 21.2 microalbuminuria, overt
proteinuria
P-DSPN: age, HbA1c,
lower HDL-c, overt
proteinuria
Type 2, Qatar [84] 1095 DN ≥ 4 34.5 P-DSPN 13.6 P-DSPN VPT, smoking, obesity,
P-DSPN (H) Non-P-­DSPN 57.5 female sex, DM duration
8.2 Non-P-­
DSPN 52.6
(continued)
17
18
Table 2 (continued)
DM duration Mean age
Study site Participants (N) Neuropathy diagnosis Prevalence (%) (mean, year) (year) Risk factors/markers
Type 1 and 2, UK [90] 350 VPT, NSS, NDS, Pain symptoms score 16.2 NA 63 NA
P-DSPN (P) ≥3 and neuropathic pain in legs ≥1
year
Type 1 + 2, England [91] T1 1338 NSS ≥ 5 and NDS ≥ 3 T1 13.4 T1 17 T1 37.6 Risk for painful
P-DSPN (P) T2 14,206 T2 21.5 T2 4 T2 63.6 symptoms: T2D, female
Overall 21 gender, South Asian
ethnicity
Type 1 + 2, Germany T1 126 DSPN: ≥1 of following: T1 44.3 of NA T1 59.5 T2 DSPN + P-DSPN:
P-DSPN (P) PROTECT study T2 943 monofilament, temperature sensation, which T2 70.0 higher and lower BMI
[85] VPT P-DSPN 54.8
P-DPSN: DSPN and pain/burning in T2 55.3 of
feet which
P-DSPN 61.7
Type 1 + 2, Belgium [7] T1 344 DSPN: Monofilament and/or pinprick T1: T1 16.5 T1 45.9 Nephropathy, obesity,
P-DSPN (H) T2 767 Painful-DSPN: DSPN and DN4 >4 DSPN 25.6 T2 11.0 T2 63.6 low HDL-c, high
P-DSPN 5.8 triglycerides
T2:
DSPN 50.8
P-DSPN 17.9
Type 1 + 2, Libya [92] T1 + T2 450 S-LANSS ≥ 12 42.2 15 50.6 DM duration, smoking
P-DSPN (H) (men), obesity, fasting
Glc
BMI body mass index, BP blood pressure, CDT cooling detection threshold, CON conventional treatment, CVD cardiovascular disease, DB deep breathing, DKA diabetic ketoaci-
dosis, DM duration diabetes mellitus duration, DN4 Douleur Neuropathique 4 Questions, ECS electrochemical skin conductance, Glc glucose, H Hospital/out-patient based,
HbA1c hemoglobin A1c, HDL/LDL/total-c high density lipoprotein/low density lipoprotein/total chlolesterol, IHD ischemic heart disease, INT intensive treatment, MI myocar-
dial infarction, MNSI examination Michigan neuropathy screening instrument examination, MNSIq Michigan neuropathy screening instrument questionnaire, NCS nerve conduc-
tion studies, NNS and NDS Neuropathy symptom score and Neuropathy disability score (Young et al. [74]), P primary care/community-based, PAD peripheral arterial disease,
S-LANSS self-report Leeds Assessment of Neuropathic Symptoms and Signs (S-LANSS) pain scale, u-ACR urin albumin-creatinine ratio, VAL Valsalva’s manoeuvre, VPT
vibration perception threshold
C. S. Hansen et al.
The Epidemiology of Diabetic Neuropathy 19

Recently, lower P-DSPN prevalences have neurological deterioration is associated with


been reported in the Danish DD2 cohort of increasing age [17] and the risk factor profile of
recently diagnosed T2D.  Confirmed P-DSPN participants may change over time. Furthermore,
was estimated to be present in only 5.4% of the nerve fibre dysfunction may be partly reversible
cohort of more than 5500 individuals when eval- [17, 99–101], further complicating interpretation
uated clinically in a sub-sample of 389 individu- of longitudinal studies of DSPN.
als [89]. By contrast, P-DSPN prevalences from A long-term clinic-based study published in
two hospital-based studies in Qatar and Libya the 1970s followed over 4000 people with T1D
report high prevalences of 34.4% and 42.2%, and T2D for 25 years and found a cumulative
respectively. The latter study was, however, based DSPN incidence of 50% [102–104]. Even though
solely on self-reported and self-evaluated neuro- some aspects of the methodology can be ques-
pathic pain [84, 92]. However, prevalences simi- tioned, the size of the study is noteworthy. DSPN
lar to the ones reported in community-based incidence has since been evaluated in more recent
samples are also found in out-patient settings [7]. study cohorts. These are summarised in the fol-
Reports characterising both DSPN and P-DSPN lowing paragraph.
estimate that approximately 20–40% of people In newly diagnosed diabetes, a small Finnish
with DSPN have neuropathic pain [7, 77, 78, 89]. cohort of people with T2D showed an increase
There is a large disparity between painful from a baseline prevalence of 8.3% to 41.9%
symptoms and clinical signs of DSPN and little is over 10 years when defining DSPN using NCS
known about the natural history of P-DSPN [5, 6, and symptoms [31]. The UKPDS cohort of newly
91, 93, 94]. There are few longitudinal studies of diagnosed T2D evaluated DSPN by VPT and
P-DSPN and most are small. These provide con- found that the prevalence increased from approx-
flicting results, indicating both spontaneous pain imately 11–30% over 12 years, irrespective of
improvement and remission over time [95, 96] as treatment arm [45]. In comparison, the
well as the opposite finding [95, 97]. As recent ADDITION-Denmark cohort of screen-detected
reviews report the duration and severity of diabe- T2D report a cumulative incidence of only 10%
tes, and the severity of neuropathy, as risk factors over 13 years of follow-up [46]. In contrast, the
for P-DSPN, pain remission seems unlikely [6, Bypass Angioplasty Revascularization
83]. In conclusion, approximately 15% of people Investigation 2 Diabetes (BARI 2D) trial of more
with diabetes have P-DSPN but estimates vary than 2,000 people with longstanding T2D and
and may be as high as 40%. confirmed coronary artery disease, showed a
DSPN prevalence of 50% at baseline and a cumu-
lative incidence over 4 years of 66–72% when
2.4 Incidence of DSPN defining DSPN using the MNSI examination
[105]. The ACCORD trial similarly defined
Although reported prevalences of DSPN in T1D DSPN using the MNSI examination. Although
and T2D are similar, the incidence of DSPN is the trial was closed earlier than planned due to
estimated to be higher in T2D (6.1 per 100 higher mortality in the intensive treatment arm,
person-­years) than in T1D (2.8 per 100 person-­ intensive treatment (INT) decreased the risk of
years). This is probably due to differences in the DSPN compared to conventional care (CON).
underlying pathophysiology and populations Baseline prevalences of 42% (CON) and 43%
characteristics of T1D and T2D [1, 6, 17, 98]. (INT) increased to 59% (CON) and 56% (INT)
The slow progression of DSPN complicates after 5 years [70]. Two other smaller longitudinal
investigation of incident DSPN and highlights studies of T2D report an annual incidence of
the importance of testing methodology. Different 6.1% when evaluating DSPN with both symp-
nerve fibre populations may be affected at differ- toms and signs [106] and 20% over 2.5 years
ent rates and minor changes may be undetected when evaluating DSPN by monofilament [62].
due to low sensitivity of tests. In addition, natural More recently, an Indian study of T2D reported a
20 C. S. Hansen et al.

4-year incidence of 28.4% evaluated using VPT 2.5 Risk Factors for DSPN
[107]. and P-DSPN
The landmark studies of DCCT/EDIC provide
detailed information on the incidence of DSPN A long list of risk factors has been associated
[21]. Incidence of confirmed DSPN was 6% and with DSPN.  Risk factors for DPSN in the
14% in the intensive and the conventional treat- reviewed studies are summarised in Tables 1 and
ment arms, respectively, at DCCT closeout after 2 and additional studies are referenced to in the
approximately 5 years. The incidence in the following paragraph.
EDIC cohort at year 13/14 was 22% (INT) and The duration of diabetes and level of hyper-
28% (CON), respectively, and the prevalence was glycaemia (HbA1c) are the two most important
25% (INT) and 35% (CON) [58, 59]. Underlying risk factors for DSPN [6, 17, 57, 111]. The
NCS abnormalities were also more prevalent in importance of hyperglycaemia has been docu-
the CON group and consistently higher than the mented in both T1D and T2D [27, 28, 31, 33, 51,
prevalence of confirmed or clinical DSPN in both 55, 57, 62, 112] and the effect of strict glycaemic
the INT and CON groups [59]. control has been investigated in several studies
The observational Epidemiology of Diabetes [45, 58, 59, 70, 113, 114]. Optimising glycaemic
Complications (EDC) study of childhood-onset control in T1D delays development of DSPN and
T1D reported a 15% prevalence of DSPN at the slows down the progression of DSPN, whereas
6-year follow-up in participants free from the effect of glucose control is less clear in T2D
DSPN at baseline, corresponding to an inci- [115]. More recently, obesity, including measures
dence of 2.8 per 100 person-years [108]. At 12 of weight, waist circumference, and BMI, have
years, the incidence per 100 person-years was emerged as important risk factors of DSPN [15–
1.2 for people with a T1D duration <20 years 17, 46, 48, 111, 116]. Other components of the
and 3.8 for a T1D duration of 20–30 years metabolic syndrome, such as hypertension, insu-
[109]. The EURODIAB study for risk factors lin resistance, and dyslipidemia, also seem to
for the development of DSPN followed over increase the risk of DSPN [6, 16, 17, 57, 111,
1000 T1D patients in multiple centres over 116]. Importantly, such factors are modifiable.
Europe for 7.3 years and found a cumulative Other independent risk factors for DSPN are
incidence of 23.5% using symptoms and signs smoking, alcohol, increased height (probably as a
[56, 57]. More recently, a population-­ based proxy for nerve fibre length), and older age [17,
cohort of over 4000 people with newly diag- 57, 111]. Alcohol consumption is also regarded as
nosed T1D from Taiwan reported of a rather a separate cause of peripheral neuropathy, but the
low cumulative incidence of 23.7% over 12 distinction between alcohol and diabetes as causal
years [110]. However, the participants were fol- risk factors for DSPN is difficult. The effect of
lowed from the time of T1D diagnosis and the age on DSPN is controversial since age per se
study was based on registries where the detec- causes progressive neurological deterioration and
tion of incident DSPN relied on a registered although age has been widely reported as an inde-
DSPN diagnosis during follow-up. pendent risk factor for DSPN, there are also stud-
In summary, the incidence of DSPN is gener- ies that show the opposite effect [17, 111]. Finally,
ally higher in T2D than T1D. However, there are recent large national cohorts of T1D in the USA
exceptions, specifically lower than usual inci- and Scotland indicate that socioeconomic disad-
dence of DSPN in T2D cohorts early in their dis- vantage may be an additional independent risk
ease trajectory. No studies have investigated factor for the development of DSPN [27, 28].
incidence rate ratios. Thus, no data is available on Independent risk factors for the development of
temporal changes in incidence of DSPN. In other P-DSPN have not been studied to the same extent
words, it is not possible to assess if the incidence as risk factors for DSPN. However, any risk factor
of DSPN follows the decline as for other diabetic for diabetes or DSPN is likely to be a risk factor
complications. for P-DSPN and clear similarities with the known
The Epidemiology of Diabetic Neuropathy 21

DSPN risk factors were detected in a recent review rons, or both, in the peripheral nervous system.
of the topic [83]. Specifically, a higher prevalence This affects various organ systems, eliciting man-
of pain has been reported in patients with more ifestations including orthostatic hypotension,
severe DSPN and female sex has been shown to be silent myocardial ischemia, resting tachycardia,
a risk factor for P-DSPN [6]. hypoglycaemia unawareness, obstipation, diar-
rhoea, faecal incontinence, urine retention, uri-
nary tract infections, sudomotor dysfunction, and
2.6 DSPN Complications sexual dysfunction. The most prevalent form of
autonomic neuropathy is cardiovascular auto-
Besides the complicating neuropathic pain, nomic neuropathy (CAN), which will be
DSPN increases the risk of foot ulcers, Charcot addressed in detail below.
neuroarthropathy and amputations [1, 3]. The
lifetime risk for developing a diabetic foot ulcer
during the course of diabetes is 25%, with 3.1 Cardiovascular Autonomic
approximately 5% requiring amputation [117]. Neuropathy
These late complications are also predictors of
mortality [1, 118]. As early identification and Cardiovascular autonomic neuropathy (CAN) is
treatment of ulcers can delay or prevent adverse defined as: “A damage to the autonomic nerve
outcomes, a comprehensive annual foot evalua- fibres that innervate the heart and blood vessels,
tion is recommended, with more frequent evalua- resulting in abnormalities in heart rate control
tions for patients with sensory loss or previous and vascular dynamics” [121].
ulceration or amputation [1, 8]. It is recom- CAN is an overlooked serious diabetic compli-
mended to educate patients in preventative foot cation associated with increased risk of morbidity
self-care and apply a multidisciplinary approach and mortality [121]. This may be due to the lack
to treating foot ulcers and high-risk feet [8]. of symptoms from early detrimental changes to
DSPN also contributes to increased risk of late and severe stages [122]. CAN symptoms are
falls and fractures through loss of sensation, pro- vague and can manifest as resting tachycardia
prioception, and temperature discrimination, as [123] and dizziness or faintness when changing
well as pain and muscle weakness, all leading to position from lying or sitting to standing [124].
increased unsteadiness [1]. Additionally, DSPN Additionally, the lack of testing capacity for CAN
negatively affects quality of life and increases the may be a cause of missing epidemiological data.
risk of depression and anxiety [1, 17]. Therefore, Changes in cardiac autonomic activities are
evaluation of the risk of falls and the effect of present even in early pre-diabetes [125]. The
diabetic neuropathy on quality of life are recom- severity of CAN increases during lifetime expo-
mended [1]. Importantly, as microvascular com- sure to manifest diabetes [121]. These abnormali-
plications linked to diabetes tend to cluster, ties can be reflected in reduced heart rate
screening, and handling of diabetic nephropathy variability (HRV) and abnormal responses to car-
and retinopathy must be kept in mind [8, 119]. diovascular autonomic reflex tests (CARTs); all
Lastly, besides the increased mortality associated signs of CAN [122].
with foot ulcers and amputations, DSPN itself Throughout the latest decades several diag-
may be a predictor of mortality [1, 118, 120]. nostic tools have been applied to assess the prev-
alence and incidence of CAN.  Adhering to
current international recommendations [1, 122]
3 Diabetic Autonomic we present a summary of studies where CARTs
Neuropathy have been used to diagnose CAN.  Acceptable
methods to assess CARTs include changes in
Autonomic neuropathy is a neurological compli- heart rate and blood pressure in response to active
cation that covers a wide range of detrimental tests. The recommended active tests include (1)
changes to parasympathetic or sympathetic neu- the measurement of heart rate change when
22 C. S. Hansen et al.

changing position from lying to standing, (2) dur- 3.2 Prevalence of CAN
ing deep breathing, (3) and the Valsalva manoeu-
vre, and (4) the assessment of blood pressure The prevalence of diabetic CAN has not been
change in response to standing (orthostatic hypo- investigated as thoroughly as the prevalence of
tension test). Several sets of normative values DSPN. Early studies such as the US Framingham
have been presented for heart rate CARTs [122]. Heart study performed in the 1980s estimated the
However, there is no consensus in terms of which prevalence of CAN using analyses of 2-hour
set of normative values to use to diagnose ECG recordings. CAN measures were found to
CAN. This, in turn, affect the estimates of preva- be decreased in people with diabetes, however,
lence. Furthermore, the number of pathological prevalence rates were not presented [130]. Later
tests used to diagnose CAN does vary. Most studies addressing the prevalence of CAN using
researchers categorise early CAN as the presence HRV and CARTs have been performed. All
of one pathological test and manifest CAN as selected references addressing the prevalence of
more than one pathological test. However, vari- CAN are summarised in Table 3.
ous other scoring systems exist, hampering uni-
form estimates of CAN prevalence. 3.2.1 Youth
In addition to CARTs, we have included stud- Autonomic dysfunction has not been investigated
ies of significance where either a combination of thoroughly in young people with diabetes and
CARTs and resting HRV was assessed or where only a few studies have applied CARTs to esti-
HRV was the only outcome. Early changes in mate CAN prevalence in youth. It is evident,
measures of CAN are seen in indices of however, that autonomic dysfunction is prevalent
HRV. Adverse changes in HRV have been found in children and adolescent people with T1D and
in non-diabetic, pre-diabetic, and diabetic indi- T2D.  Autonomic dysfunction has been investi-
viduals [73, 126–131], underlining that changes gated in cohorts of young T1D patients with a
are not specific to diabetes, but can be seen even heterogenic range of methods such as pupil size
before the disease is diagnosed. In many cases, determination, postural systolic blood pressure
lack of normative reference values for HRV response, hand grip test, and baroreflex [162].
makes comparisons to other studies difficult. It Due to the different assessment modalities, CAN
has also been suggested that HRV measures are prevalence was reported varying from 3% to
prone to moderate reproducibility [132, 133]. 75%. These studies will not be addressed
In geographic regions where studies are further.
scarce, we have included studies where cold pres- Larger studies have applied either HRV or
sure tests and handgrip tests have been utilised. CARTs to investigate the prevalence of
Neither test has been endorsed as a recommended CAN. Using CARTs and handgrip tests research-
diagnostic tests for CAN. The handgrip test has a ers found a prevalence of 8% in 221 Chinese chil-
poor association with CARTs [134]. It is also dren with a mean age of 9 years. In the
worth mentioning the assessment of cardiovascu- SEARCH-study performed in the US, CAN was
lar autonomic dysfunction by baroreflex sensitiv- defined as three or more out of five HRV indices
ity. This test describes the relationship between compared to healthy controls. The mean age of
resting blood pressure and heart rate as a measure participants was 18 years. The prevalence was
of autonomic function [122, 135]. However, this estimated to be 18% in this cohort of 1646 pri-
method has not been applied in larger epidemio- marily T1D children and adolescents [159]. A
logical studies. Advanced cardiac imaging has recent hospital-based Danish study assessed
been utilised in few studies addressing cardiac CAN by CARTs and showed a prevalence rate of
sympathetic function [136, 137], but these tests definitive CAN of 9% in a cohort of 156 ran-
are expensive and time consuming and have not domly recruited T1D patients with a mean age of
been applied to assess the prevalence of CAN on 22 years [61]. Prevalence rates vary substantially
a population-based scale. from study to study. The EURODIAB study,
The Epidemiology of Diabetic Neuropathy 23

Table 3  Prevalence of CAN in type 1 and type 2 diabetes


Prevalence (%) Diabetes
(H = Hospital, duration Mean
Diabetes Participants CAN P = primary (mean, age
type Study (N) measures care) year) (year) Risk factors/markers
Type 1 USA [138] 168 CARTs 37.5%(H) 20 29 Age, diabetes
(DB) duration, HbA1c,,
diastolic blood
pressure, LDL, HDL,
triglycerides,
hypertension,
retinopathy
Type 1 Europe [139] 1086 CARTs (LS) 32% (H) 1–7 15–29 age, diabetes
35% 8–14 30–44 duration, HbA1c,,
52% 45–60 45–60 diastolic blood
pressure, HDL,
albuminuria,
smoking, cholesterol/
HDL-cholesterol,
triglycerides
Type 1 Europe [140] NA CARTs NA NA NA NA
(LS, OH)
Type 1 Brazil [141] 151 CARTs (LS, 11.3% 16.3 33.4 Age, diabetes
DB, VM, (19.2%. early duration, HbA1c,
OH) CAN) (H) weight, hypertension,
creatinine, LDL,
eGFR, albuminuria
Type 1 USA [59, 620a R-R 5%a (H) 5.5a 25.5a NA
142] 570b variation, 10%b 11.9b 33.0b
594c valsalva 35%c 26.1 47.0c
ration, OH
Type 1 Denmark 156 CARTs (LS, Early CAN 11.3 22 NA
[61] DB, VM) 28.1% (P)
Definite CAN
9%
Type 1 Egypt [143] 60 CARTs (LS, Early CAN 7.6 15.1 HbA1c
DB, VM, 54.5 (H)
OH, HG, Definite 27.7
cold Severe 18.2
pressure) (H)
Type 1 Korea [144] 95 CARTs (LS, Early CAN 13.1 22.2 NA
DB, VM, 10.5(H)
OH) (H) Definite 2.1
Type 2 Germany 95 CARTs (LS, <1 (H) 0 49.7 NA
[145] DB, VM) (screen-­
(H) detected
dm)
Type 2 Germany HRV Kan ikke få
[146] fat I artikel
Type 2 India [147] 145 CARTs (LS, 44.2(H) 0 (screen 48 NA
DB, VM, detected
OH, HG, dm)
cold
pressure)
(continued)
24 C. S. Hansen et al.

Table 3 (continued)
Prevalence (%) Diabetes
(H = Hospital, duration Mean
Diabetes Participants CAN P = primary (mean, age
type Study (N) measures care) year) (year) Risk factors/markers
Type 2 Pakistan 207 HR, OH, 36.7 (H) NA 54.6 NA
[148] Handgrip,
etc.
Type 2 USA [149] 8135 HRV, QTI 3.5% (H) 10.8 62 HbA1c,, diastolic
blood pressure, BMI,
and triglycerides
Type 2 Italy [150] 557 CARTs (LS, 1.8%(H) <6 58.3 BMI, HbA1c,
DB, VM, (15.3%). months AH-drugs
OH) Early CAN)
(NA)
Type 2 Denmark 469 CARTs (LS, 13.4% (H) 10.0 59.0 Vitamin B12
[151] DB, VM)
Type 2 Korea [152] 3199 CARTs (LS, 23.7 (H) 5.5 59 Age, diabetes
DB, VM) duration,
(H) hypertension, eGFR,
retinopathy,
nephropathy, ApoA-1
Type 2 Denmark 443 CARTs (LS, 15.1 (P) (H) 12.8 70.3 Age, BMI, HbA1c,
[153] DB, VM) weight, diastolic
blood pressure,
Triglycerides
Type 2 Denmark 777 CARTs (LS, 9.0 (P) 6.3 65.0 Diabetes duration,
[153] DB, VM) BMI, HbA1c, weight,
waist circumference,
LDL, Triglycerides,
eGFR, albuminuria
Type 2 Korea [154] 1458 CARTs (LS, 55.7 (H) 8.2 58.1 NA
DB, VM)
(H)
Type 2 India [155] 100 CARTs (LS, Early CAN 25 9.0 53.3 Diabetes duration,
DB, VM, (H) creatinine, eGFR, age
OH) (H) Definite 24
Type 2 USA [156] 4629 HRV 9.7% (H) 4.3 59.7 Age, African-­
(10sek) American ethnicity,
(2of2) BMI, diastolic BP,
triglycerides,
Smoking
Type 1 Germany, T1dm: CARTs (LS, t1dm: 16.8% NA T1dm: NA
and 2 Switzerland, 647 DB, VM) (H) 11–69
Austria [73] T2dm: HRV (8.5% early T2dm:
524 CAN) 16–72
t2dm:
22.1%
(12.2% early
CAN) (H)
Type 1 France [157] T1dm: CARTs (LS, t1dm: 23.0% T1dm: T1dm: Age, Diabetes
and 2 245 DB, VM) t2dm:15.5% 10.0 36.1 duration, HbA1c,
T2dm: (H) T2dm: T2dm: BMI, systolic/
151 6.3 45.5 diastolic blood
pressure, albuminuria
The Epidemiology of Diabetic Neuropathy 25

Table 3 (continued)
Prevalence (%) Diabetes
(H = Hospital, duration Mean
Diabetes Participants CAN P = primary (mean, age
type Study (N) measures care) year) (year) Risk factors/markers
Type 1 USA [158] T1dm: 83 CARTs (LS, t1dm: 10.8% T1dm: T1dm: NA
and 2 T2dm: DB, VM, t2dm:16.2% 24.3 50.9
148 OH) (NA) T2dm: T2dm:
Symptoms 15.3 64.1
Type 1 USA [159] 1646 HRV (≥3 of 12%(t1dm) 8 18 HbA1c, BP,
and 2 5) (H) triglycerides (t1dm),
17%(t2dm) albuminuria
Type 1 Uganda [160] 299 CARTs Early CAN 5.8 50.1 Age, diabetes
and 2 (DB, VM, 29.4(H) duration, retinopathy
OH, Definite 20.4
HRHGa) Severe 2.3
Type 1 China [161] 73 (t1dm) CARTs (LS, 61.6(t1dm) NA NA Age, diabetes
and 2 1974 DB, VM, (H) duration
(t2dm) OH) (H) 62.6(t1dm)
LS heart response to lying to standing position, DB heart rate response to deep breathing, VM heart rate response to
Valsalva manoeuvre, OH orthostatic hypotension test, HG blood pressure response to hand grip, HR resting heart rate,
HRV heart rate variability, SM sudomotor function, HDL high density lipoprotein, LDL low density lipoprotein, eGFR
estimated glomerular filtration rate, BMI body mass index
a
DCCT baseline (control group)
b
DCCT close-out (control group)
c
EDIC 13–14 year follow-up (control group)

where CAN was assessed only by heart rate a prevalence of 16.8% based on CARTs and
response to deep breathing, found a prevalence of HRV measures.
32% in T1D patients in the youngest age group Later hospital-based studies have reported a
(on average 22 years of age) [139]. This preva- similar prevalence of CAN based on CARTs in
lence could be interpreted as a marker of early T1D. In Brazil, the prevalence of CAN is 11.3%
CAN and is comparable with the prevalence of in patients with a mean age of 33.4 years [141].
early CAN in the Danish study where a preva- In the DCCT/EDIC study, participants at DCCT
lence of 28.1% was reported. close-out aged approximately 33 years had a
Thus, CAN is a significant complication in CAN prevalence of 10% when assessed by HRV,
children and adolescents. Prevalence rates in Valsalva heart rate test, and orthostatic hypoten-
children vary from 5% to 8% and in younger sion [59, 142]. In the EURODIAB study a much
adults from about 9% to 18% depending on the higher prevalence of CAN was reported in simi-
methods applied. The prevalence of early CAN lar age groups. Here a prevalence of 35% was
may be about 30%. seen in patients in the 30–44-year age range.
CAN was assessed only by heart rate change to
3.2.2 Type 1 Diabetes deep breathing, possibly constituting both early
The majority of CAN prevalence studies have and definite CAN.  In 2018 definite CAN was
been performed in T2D patients. However, larger reported tin 27.1% of patients in a smaller
studies in T1D cohorts or mixed T1D and T2D Egyptian cohort, where assessments were per-
patients have also been conducted. formed using CARTs, hand grip, and cold pres-
Only few studies of significant size have sures test [143]. Similarly, a high prevalence of
been performed prior to 2000. The European 23% was found in a French multicentre study of
multicentre study DIACAN was conducted in 245 patients with a mean age of 36 years. In 2019
the early 1990s in several hospital-based diabe- an unprecedented prevalence of 61.2% was
tes centres. Here patients aged 11–69 years had observed in T1D [161] patients in a hospital-­
26 C. S. Hansen et al.

based Chinese cohort. However, CAN was was found; when using HRV to diagnose CAN,
assessed as a composite of an array of symptoms only 9.7% had CAN [156].
and objective measures, which may contribute to Few studies have examined CAN in patients
the high estimate. with mean diabetes durations of approximately
In conclusion only few significant studies are 10–15 years. Danish studies indicate a CAN
available on CAN prevalence in adults with T1D prevalence (≥2 pathological CARTS) of about
using recommended CARTs. The prevalence var- 15%. The ADDITION-Denmark 13-year follow-
ies from approximately 10%–27%. Regional dif- ­up study showed that 15.1% had definitive CAN
ferences may apply, but heterogeneous [153] and another study of randomly recruited
assessment methods hamper robust patients showed a prevalence of 13.4% [151].
comparisons. American data seems to mirror findings from
European studies. Using a complex scoring sys-
3.2.3 Type 2 diabetes tem of CARTs and symptoms, CAN prevalence
A few noteworthy studies of European origin has been reported to be approximately 16% in
were performed in the 1990s. In these studies patients with a diabetes duration of 15.3 years
CAN was assessed by CARTs with [163] or with- [158]. Only 3.5% of participants in the ACCORD
out [145] a handgrip test. CAN was present in study had CAN, however, here CAN was assessed
<1% and 8.1% of patients, respectively. The first by HRV and QT-interval of ECG recordings
study was an investigation of newly diagnosed [149]. Higher prevalence has been reported in
patients. With increasing age, a higher prevalence Asian studies. Indian data suggests that more
has been reported by others, where estimates of patients have CAN when recommended CARTs
definite CAN reached 22.1% [73]. are utilised. In a smaller cohort of 100 patients
Since 2000, definite CAN has been explored 24% had definite CAN [155]. When diagnosing
in several large cohorts. Studies investigating CAN as one or more pathological CARTs, a
patients with short diabetes duration indicate a CAN prevalence of 55.7% was found in a large
low prevalence of CAN in early T2D. Using rec- (N = 1458) Korean hospital-based study [154]. In
ommended CARTs and the orthostatic hypoten- 2019 an unprecedented prevalence of 62.6 % was
sion test, Italian researchers reported a prevalence observed in T2D patients in a hospital-based
of definite CAN of 1.8% and early CAN of 15% Chinese cohort [161]. However, CAN was
in patients with a diabetes duration of less than assessed using a composite array of symptoms
six months [150]. Hospital-based Indian data of and objective measures, which may contribute to
screen-detected T2D indicate regional differ- the high estimate. A Pakistani study of 200 T2D
ences. Defining CAN as one or more pathologi- patients showed a prevalence of 36.7% (mean
cal test of six tests, 44.2% were diagnosed with age 54.6 years), but diabetes duration was not
CAN [147]. Here, no discrimination was applied reported and CAN was assessed by heart rate,
between early and definite CAN. QTc-interval, Handgrip, and orthostatic hypoten-
With longer diabetes duration higher CAN sion tests [148], making the study difficult to
prevalence has been reported. Diabetes duration compare with others.
of approximately 5 years have been associated In conclusion, CAN prevalence in T2D varies
with definite CAN assessed by CARTs (≥2 path- by geography, with lower rates in western coun-
ological CARTS), with a prevalence between 9% tries. The prevalence in early T2D rages from
and 24% (Table 3). Geographical differences are <1% to a few percent increasing to 15–22% in
apparent. Findings range from 9% in the western cohorts and possibly even higher (24–
ADDITION-Denmark study [153] and 15.5% in 37%) in non-western populations at later stages
a French study [153] to approximately 25% in of the disease. No apparent changes in prevalence
studies from Uganda [160] and Korea [152]. In have been observed from the 1990s to the
the GRADE study a relative low CAN prevalence present.
The Epidemiology of Diabetic Neuropathy 27

3.3 Incidence of CAN In the recent ADDITION-Denmark study,


which included patients with screen-detected dia-
A few larger longitudinal study cohorts have betes in primary care, low progression rates of
been established. These studies are either obser- CAN were reported. Participants were screened
vational cohorts or randomised controlled trials. from 2001 and 2006  in primary care. The preva-
For the latter study design valuable information lence of confirmed CAN (≥2 pathological CARTS)
on the natural history of CAN can be derived increased from 9.0% to 15.1% with an annual inci-
from the control groups. dence of 1.8% between the 6- and 13-year follow-
In the observational multi-national up of the cohort [153]. The ADDITION-Denmark
EURODIAB study, which began in 1989, 956 study also evaluated the flow of CAN stages in par-
T1D patients were examined. CAN was diag- ticipants re-examined for CAN (Fig. 1). There was
nosed by either having impaired heart rate or a progressive change in CAN status from 6- to
blood pressures response to standing. In total, 13-year follow-up, although the phenotype of CAN
17% of participants developed CAN in the 7.3-­ was heterogeneous and improvements were seen in
year follow-up period, a mean annual increase of all categories of CAN, suggesting CAN to have an
approximately 2.3%. This was equivalent to an element of reversibility.
incidence rate of 23.4 per 1000 person-years of In summary, only a very few longitudinal stud-
follow-up [140]. In the DCCT/EDIC study a ran- ies with a significant sample-size have been per-
domised placebo-controlled trial with interven- formed to accurately assess CAN incidence.
tion free follow-up, 5% of participants in the Annual progression rates seem similar in both T1D
control group had CAN at baseline (defined by and T2D patients at approximately 2%. No studies
having either abnormal HRV, heart rate response have been performed on population-based cohorts
to Valsalva or orthostatic hypotension). The study that allow for assessment of incidences rate ratios
enrolled patients from 1983 to 1989. In the for- for CAN. Therefore, as for DSPN it is not possible
mer control group 35% of participants had CAN to document a reduction in the incidence of CAN
after 14 years of EDIC follow-up [59, 142]. This over time that can mirror the improvements of pre-
approximates an annual incidence of 2.1%. venting other diabetic complications.

Fig. 1  The flow of participants between clinical groups ries; No CAN (green), early CAN (yellow) or manifest
of CAN: ADDITION-Denmark. Number of participants CAN (red) from the 6- to the 13-year follow-up [153]
and changes in their CAN status among defined catego- (used with permission from Diabetes Care)
28 C. S. Hansen et al.

3.4 Risk Factors for CAN tile dysfunction (ED) and is more prevalent in
diabetes patients compared to men without dia-
Several risk factors are associated with diabetic betes. This may be associated with autonomic
CAN in cross-sectional investigations, e.g. diabe- dysfunction. However, a common cause of ED is
tes duration, higher HbA1c, HDL, triglycerides, atherosclerosis. So far, no studies have published
higher blood pressure and BMI.  Nephropathy, data to discriminate between these two causes of
low vitamin B12, DPSN, and retinopathy have ED making it impossible to investigate the preva-
also been found to be associated with CAN lence of autonomic ED. Similarly, female sexual
(Table 3). Few studies have addressed risk factors dysfunction is more prevalent in people with dia-
for progression to CAN. The EURODIAB study betes. Again, no studies have been able to assess
showed that elevated HbA1c, hypertension, the prevalence of female sexual dysfunction
DSPN, and retinopathy predict the risk of devel- attributed to autonomic neuropathy.
oping CAN [140]. The ADDITION-Denmark Urinary incontinence and bladder dysfunction
study did not reveal any statistically significant are linked to the presence of diabetic neuropathy
risk factors for incident CAN.  However, inter- in both men and women [171]. However, the
vention studies have demonstrated that multifac- prevalence of bladder dysfunction due to auto-
torial intervention [164] and intensive nomic dysfunction has not been investigated.
glucose-lowering treatment [21, 165, 166] can Autonomic dysfunction of the skin is seen as
reduce progression to manifest CAN. both decreased (hypohidrosis) and increased
(hyperhidrosis) sweat production. The preva-
lence of these dysfunctions are unknown.
3.5 Other Forms of Autonomic
Neuropathy
4 Other Forms of Diabetic
As mentioned above, other organ systems can be Neuropathy
affected by autonomic neuropathy.
Gastrointestinal autonomic neuropathy is less Diabetic mononeuropathies and radiculopathies
common and can be subdivided into gastropare- (or polyradiculopathies) with focal presentations
ses affecting gastric emptying and diabetic enter- in the innervation area of the affected nerve or
opathy. Epidemiological data on these nerve root are atypical forms of diabetic neuropa-
complications are scarce and mainly based on thy and differ from DSPN in onset, course, and
symptoms. It is estimated that 1–50% [167–169] manifestations [1, 3, 10].
of patients with diabetes have symptoms of gas- Mononeuropathies due to compression occur
troparesis, a complication causing dysmotility of more commonly in people with diabetes and
the stomach. Gastroparesis is associated with most often involve the median, ulnar, radial, and
poor glycaemic control, higher mean levels of common peroneal nerves [10, 172]. However, the
HbA1c, and risk of severe hypoglycaemia. explanation for the predisposition of nerve injury
Furthermore, diabetic gastroparesis is associated in diabetic mononeuropathies is considered mul-
with poor drug absorption, malnutrition, and tifactorial. Mononeuropathies in people with dia-
reduced quality of life due to gastrointestinal betes may not be exclusively due to entrapment
symptoms [170]. Diabetic enteropathy, where [172]. This is exemplified by studies showing
nerves innervation the intestines are affected, is that over 20% of patients investigated for median
less well studied. No studies have assessed the mononeuropathy fulfilled NCS criteria for carpal
prevalence of diabetic enteropathy using objec- tunnel syndrome, even though they were asymp-
tive measures. tomptomatic [51, 172, 173]. Cranial neuropathies
Autonomic neuropathy may manifest as sex- are more rare, present acutely, and can affect up
ual dysfunction and bladder dysfunction. Sexual to 1% of people with diabetes, primarily involv-
dysfunction in men is typically observed as erec- ing the 3rd, 4th, 6th, and 7th cranial nerve [1, 10].
The Epidemiology of Diabetic Neuropathy 29

Diabetic radiculoplexus neuropathy (DRPN) 7. Van Acker K, et al. Prevalence and impact on quality
of life of peripheral neuropathy with or without neu-
or polyradiculoneuropathy presents with cervi- ropathic pain in type 1 and type 2 diabetic patients
cal, thoracic, and/or lumbosacral involvement. attending hospital outpatients clinics. Diabetes
Lumbosacral DRPN (diabetic amyotrophy) is the Metab. 2009;35:206–13. https://doi.org/10.1016/j.
most common condition and is estimated to have diabet.2008.11.004.
8. American Diabetes Association. Microvascular com-
a lifelong incidence of 1% in people with diabe- plications and foot care: standards of medical care in
tes [172, 174]. Affected people are typically diabetes-2021. Diabetes Care. 2021;44:S151–s167.
middle-­aged or elderly and have T2D. Glycaemic https://doi.org/10.2337/dc21-­S011.
control, diabetes duration, treatment or severity 9. Dyck PJ, et al. Diabetic polyneuropathies: update on
research definition, diagnostic criteria and estima-
of DSPN are unrelated to the risk of DRPN [10, tion of severity. Diabetes. 2011;27:620–8. https://
174]. The presentation is usually abrupt with doi.org/10.1002/dmrr.1226.
severe unilateral thigh pain followed by progres- 10. Smith AG, Singleton JR.  Diabetic neuropa-
sive atrophy, weakness and weight loss. The dis- thy. Continuum. 2012;18:60–84. https://doi.
org/10.1212/01.CON.0000411568.34085.3e.
order is usually self-limiting and is considered 11. Singleton JR, Smith AG, Bromberg MB. Increased
immune-mediated [10, 172]. prevalence of impaired glucose tolerance in
Lastly, treatment induced neuropathy (TIN) is patients with painful sensory neuropathy. Diabetes
worth mentioning. TIN is considered a rare, per- Care. 2001;24:1448–53. https://doi.org/10.2337/
diacare.24.8.1448.
haps under-recognised, iatrogenic small fibre 12. Asghar O, et al. Corneal confocal microscopy detects
neuropathy, that appears after abrupt lowering of neuropathy in subjects with impaired glucose toler-
hyperglycaemia leading to a painful sensory neu- ance. Diabetes Care. 2014;37(9):2643–6. https://doi.
ropathy often with autonomic manifestations [10, org/10.2337/dc14-­0279.
13. Bongaerts BWC, et al. Older subjects with diabetes
175]. The disorder is most often seen in T1D and and prediabetes are frequently unaware of having
has a prevalence of up to 10% in patients with distal sensorimotor polyneuropathy: The KORA F4
diabetes referred for evaluation of diabetic neu- study. Diabetes Care. 2013;36:1141–6. https://doi.
ropathy to a tertiary referral centre [175]. org/10.2337/dc12-­0744.
14. Im S, Kim SR, Park JH, Kim YS, Park GY. Assessment
of the medial dorsal cutaneous, dorsal sural, and
medial plantar nerves in impaired glucose toler-
References ance and diabetic patients with normal sural and
superficial peroneal nerve responses. Diabetes Care.
1. Pop-Busui R, et al. Diabetic neuropathy: a position 2012;35:834–9. https://doi.org/10.2337/dc11-­1001.
statement by the American Diabetes Association. 15. Ziegler D, Rathmann W, Dickhaus T, Meisinger
Diabetes Care. 2017;40:136–54. https://doi. C, Mielck A. Prevalence of polyneuropathy in pre-­
org/10.2337/dc16-­2042. diabetes and diabetes is associated with abdomi-
2. Gregg EW, et al. Changes in diabetes-related com- nal obesity and macroangiopathy: the MONICA/
plications in the United States, 1990-2010. N Engl KORA Augsburg Surveys S2 and S3. Diabetes
J Med. 2014;370:1514–23. https://doi.org/10.1056/ Care. 2008;31:464–9. https://doi.org/10.2337/
NEJMoa1310799. dc07-­1796.
3. Tesfaye S, et  al. Diabetic neuropathies: update on 16. Callaghan BC, et  al. Metabolic syndrome compo-
definitions, diagnostic criteria, estimation of ­severity, nents are associated with symptomatic polyneuropa-
and treatments. Diabetes Care. 2010;33:2285–93. thy independent of glycemic status. Diabetes Care.
https://doi.org/10.2337/dc10-­1303. 2016;39:801–7. https://doi.org/10.2337/dc16-­0081.
4. Callaghan BC, Price RS, Feldman EL.  Distal 17. Ziegler D, Papanas N, Vinik AI, Shaw
symmetric polyneuropathy: a review. JAMA. JE.  Epidemiology of polyneuropathy in dia-
2015;314:2172–81. https://doi.org/10.1001/ betes and prediabetes. Handb Clin Neurol.
jama.2015.13611. 2014;126:3–22. https://doi.org/10.1016/
5. Feldman EL, Nave KA, Jensen TS, Bennett b978-­0-­444-­53480-­4.00001-­1.
DLH.  New horizons in diabetic neuropathy: 18. Malik RA, et al. Small fibre neuropathy: role in the
mechanisms, bioenergetics, and pain. Neuron. diagnosis of diabetic sensorimotor polyneuropathy.
2017;93:1296–313. https://doi.org/10.1016/j. Diabetes. 2011;27:678–84. https://doi.org/10.1002/
neuron.2017.02.005. dmrr.1222.
6. Feldman EL, et  al. Diabetic neuropathy. Nat 19. Breiner A, Lovblom LE, Perkins BA, Bril V.  Does
Rev. 2019;5:41. https://doi.org/10.1038/ the prevailing hypothesis that small-fiber dysfunc-
s41572-­019-­0092-­1. tion precedes large-fiber dysfunction apply to type 1
30 C. S. Hansen et al.

diabetic patients? Diabetes Care. 2014;37:1418–24. 31. Partanen J, et al. Natural history of peripheral neu-
https://doi.org/10.2337/dc13-­2005. ropathy in patients with non-insulin-dependent
20. England JD, et  al. Distal symmetric polyneuropa- diabetes mellitus. N Engl J Med. 1995;333:89–94.
thy: a definition for clinical research: report of the https://doi.org/10.1056/nejm199507133330203.
American Academy of Neurology, the American 32. Franklin GM, Kahn LB, Baxter J, Marshall JA,
Association of Electrodiagnostic Medicine, and Hamman RF.  Sensory neuropathy in non-insulin-­
the American Academy of Physical Medicine and dependent diabetes mellitus. The San Luis Valley
Rehabilitation. Neurology. 2005;64:199–207. Diabetes Study. Am J Epidemiol. 1990;131:633–
https://doi.org/10.1212/01.Wnl.0000149522.32823. 43. https://doi.org/10.1093/oxfordjournals.aje.
Ea. a115547.
21. The Diabetes Control and Complications Trial 33. Herman WH, et  al. Diabetes mellitus in
Research Group. The effect of intensive diabetes Egypt: glycaemic control and microvascular
therapy on the development and progression of neu- and neuropathic complications. Diabet Med.
ropathy. Ann Intern Med. 1995;122:561–8. 1998;15:1045–51. https://doi.org/10.1002/
22. Hanewinckel R, et  al. Prevalence of polyneuropa- (sici)1096-­9 136(1998120)15:12<1045::Aid-­
thy in the general middle-aged and elderly popu- dia696>3.0.Co;2-­l.
lation. Neurology. 2016;87:1892–8. https://doi. 34. Shaw JE, et  al. Diabetic neuropathy in Mauritius:
org/10.1212/wnl.0000000000003293. prevalence and risk factors. Diabetes Res Clin
23. Hanewinckel R, van Oijen M, Ikram MA, van Doorn Pract. 1998;42:131–9. https://doi.org/10.1016/
PA.  The epidemiology and risk factors of chronic s0168-­8227(98)00100-­4.
polyneuropathy. Eur J Epidemiol. 2016;31:5–20. 35. Verhoeven S, van Ballegooie E, Casparie AF. Impact
https://doi.org/10.1007/s10654-­015-­0094-­6. of late complications in type 2 diabetes in a Dutch
24. Maser RE, et  al. Epidemiological correlates of population. Diabet Med. 1991;8:435–8. https://doi.
diabetic neuropathy. Report from Pittsburgh org/10.1111/j.1464-­5491.1991.tb01627.x.
Epidemiology of Diabetes Complications Study. 36. Beghi E, Monticelli ML. Diabetic polyneuropathy in
Diabetes. 1989;38:1456–61. the elderly. Prevalence and risk factors in two geo-
25. Veglio M, Sivieri R.  Prevalence of neuropa- graphic areas of Italy. Italian General Practitioner
thy in IDDM patients in Piemonte, Italy. The Study Group (IGPSG). Acta Neurol Scand.
Neuropathy Study Group of the Italian Society for 1997;96:223–8.
the Study of Diabetes, Piemonte Affiliate. Diabetes 37. Kumar S, et al. The prevalence of foot ulceration and
Care. 1993;16:456–61. https://doi.org/10.2337/ its correlates in type 2 diabetic patients: a population-­
diacare.16.2.456. based study. Diabet Med. 1994;11:480–4. https://
26. Olsen BS, et  al. A 6-year nationwide cohort doi.org/10.1111/j.1464-­5491.1994.tb00310.x.
study of glycaemic control in young people with 38. Barbosa AP, Medina JL, Ramos EP, Barros
type 1 diabetes. Risk markers for the develop- HP.  Prevalence and risk factors of clinical diabetic
ment of retinopathy, nephropathy and neuropathy. polyneuropathy in a Portuguese primary health care
Danish Study Group of Diabetes in Childhood. J population. Diabetes Metab. 2001;27:496–502.
Diabetes Complicat. 2000;14:295–300. https://doi. 39. Mimi O, Teng CL, Chia YC. The prevalence of dia-
org/10.1016/s1056-­8727(00)00078-­7. betic peripheral neuropathy in an outpatient setting.
27. Mizokami-Stout KR, et  al. The contemporary Med J Malaysia. 2003;58:533–8.
prevalence of diabetic neuropathy in type 1 dia- 40. Al-Mahroos F, Al-Roomi K.  Diabetic neuropathy,
betes: findings from the T1D exchange. Diabetes foot ulceration, peripheral vascular disease and
Care. 2020;43:806–12. https://doi.org/10.2337/ potential risk factors among patients with diabe-
dc19-­1583. tes in Bahrain: a nationwide primary care diabetes
28. Jeyam A, et  al. Diabetic neuropathy is a substan- clinic-based study. Ann Saudi Med. 2007;27:25–31.
tial burden in people with type 1 diabetes and is https://doi.org/10.5144/0256-­4947.2007.25.
strongly associated with socioeconomic disad- 41. Pradeepa R, et al. Prevalence and risk factors for dia-
vantage: a population-­ representative study from betic neuropathy in an urban south Indian popula-
Scotland. Diabetes Care. 2020;43:734–42. https:// tion: the Chennai Urban Rural Epidemiology Study
doi.org/10.2337/dc19-­1582. (CURES-55). Diabet Med. 2008;25:407–12. https://
29. Fujimoto WY, et  al. Prevalence of complications doi.org/10.1111/j.1464-­5491.2008.02397.x.
among second-generation Japanese-American men 42. Ziegler D, Rathmann W, Dickhaus T, Meisinger
with diabetes, impaired glucose tolerance, or normal C, Mielck A. Prevalence of polyneuropathy in pre-
glucose tolerance. Diabetes. 1987;36:730–9. https:// diabetes and diabetes is associated with abdomi-
doi.org/10.2337/diab.36.6.730. nal obesity and macroangiopathy: The Monica/
30. Lehtinen JM, Uusitupa M, Siitonen O, Pyörälä Kora Augsburg surveys S2 and S3. Diabetes Care.
K.  Prevalence of neuropathy in newly diag- 2008;31:464–9. https://doi.org/10.2337/dc07-­1796.
nosed NIDDM and nondiabetic control subjects. 43. Kärvestedt L, et  al. Peripheral sensory neuropathy
Diabetes. 1989;38:1307–13. https://doi.org/10.2337/ associates with micro- or macroangiopathy: results
diab.38.10.1307. from a population-based study of type 2 diabetic
The Epidemiology of Diabetic Neuropathy 31

patients in Sweden. Diabetes Care. 2009;32:317–22. 56. Tesfaye S, et  al. Prevalence of diabetic periph-
https://doi.org/10.2337/dc08-­1250. eral neuropathy and its relation to glycaemic con-
44. Hsu WC, Yen AM, Liou HH, Wang HC, Chen trol and potential risk factors: the EURODIAB
TH.  Prevalence and risk factors of somatic and IDDM Complications Study. Diabetologia.
autonomic neuropathy in prediabetic and diabetic 1996;39:1377–84.
patients. Neuroepidemiology. 2009;33:344–9. 57. Tesfaye S, et  al. Vascular risk factors and diabetic
https://doi.org/10.1159/000254571. neuropathy. N Engl J Med. 2005;352:341–50.
45. UK Prospective Diabetes Study (UKPDS) Group. https://doi.org/10.1056/NEJMoa032782.
Intensive blood-glucose control with sulphonylureas 58. Albers JW, et  al. Effect of prior intensive insu-
or insulin compared with conventional treatment and lin treatment during the Diabetes Control and
risk of complications in patients with type 2 diabetes Complications Trial (DCCT) on peripheral neuropa-
(UKPDS 33). Lancet. 1998;352:837–53. thy in type 1 diabetes during the Epidemiology of
46. Andersen ST, et al. Risk factors for incident diabetic Diabetes Interventions and Complications (EDIC)
polyneuropathy in a cohort with screen-detected study. Diabetes Care. 2010;33:1090–6. https://doi.
type 2 diabetes followed for 13 years: ADDITION-­ org/10.2337/dc09-­1941.
Denmark. Diabetes Care. 2018;41:1068–75. https:// 59. Martin CL, Albers JW, Pop-Busui R.  Neuropathy
doi.org/10.2337/dc17-­2062. and related findings in the diabetes control and
47. Charles M, et  al. Prevalence of neuropathy and complications trial/epidemiology of diabetes inter-
peripheral arterial disease and the impact of treat- ventions and complications study. Diabetes Care.
ment in people with screen-detected type 2 dia- 2014;37:31–8. https://doi.org/10.2337/dc13-­2114.
betes: The ADDITION-Denmark study. Diabetes 60. Braffett BH, et al. Risk factors for diabetic periph-
Care. 2011;34(10):2244–9. https://doi.org/10.2337/ eral neuropathy and cardiovascular autonomic
dc11-­0903. neuropathy in the diabetes control and compli-
48. Andersen ST, et al. Risk-factor trajectories preced- cations trial/epidemiology of diabetes interven-
ing diabetic polyneuropathy: ADDITION-Denmark. tions and complications (DCCT/EDIC) study.
Diabetes Care. 2018;41:1955–62. https://doi. Diabetes. 2020;69:1000–10. https://doi.org/10.2337/
org/10.2337/dc18-­0392. db19-­1046.
49. Reis de Matos M, et  al. Distal symmetric and car- 61. Christensen MMB, et al. Prevalence of diabetic neu-
diovascular autonomic neuropathies in Brazilian ropathy in young adults with type 1 diabetes and the
individuals with type 2 diabetes followed in a pri- Association with Insulin Pump Therapy. Diabetes
mary health care unit: a cross-sectional study. Int J Technol Therap. 2018. https://doi.org/10.1089/
Environ Res Public Health. 2020;17:3232. https:// dia.2018.0249.
doi.org/10.3390/ijerph17093232. 62. Adler AI, et al. Risk factors for diabetic peripheral
50. Knuiman MW, Welborn TA, McCann VJ, Stanton sensory neuropathy. Results of the Seattle Prospective
KG, Constable IJ.  Prevalence of diabetic com- Diabetic Foot Study. Diabetes Care. 1997;20:1162–
plications in relation to risk factors. Diabetes. 7. https://doi.org/10.2337/diacare.20.7.1162.
1986;35:1332–9. https://doi.org/10.2337/ 63. Wikblad K, Smide B, Bergström A, Kessi J, Mugusi
diab.35.12.1332. F.  Outcome of clinical foot examination in rela-
51. Dyck PJ, et al. The prevalence by staged severity of tion to self-perceived health and glycaemic control
various types of diabetic neuropathy, retinopathy, in a group of urban Tanzanian diabetic patients.
and nephropathy in a population-based cohort: the Diabetes Res Clin Pract. 1997;37:185–92. https://
Rochester Diabetic Neuropathy Study. Neurology. doi.org/10.1016/s0168-­8227(97)00072-­7.
1993;43:817–24. 64. Nielsen JV.  Peripheral neuropathy, hypertension,
52. Dyck PJ, et  al. Human diabetic endoneurial sorbi- foot ulcers and amputations among Saudi Arabian
tol, fructose, and myo-inositol related to sural nerve patients with type 2 diabetes. Diabetes Res Clin
morphometry. Ann Neurol. 1980;8:590–6. https:// Pract. 1998;41:63–9. https://doi.org/10.1016/
doi.org/10.1002/ana.410080608. s0168-­8227(98)00059-­x.
53. Harris M, Eastman R, Cowie C. Symptoms of sen- 65. de Neeling JN, Beks PJ, Bertelsmann FW, Heine
sory neuropathy in adults with NIDDM in the U.S. RJ, Bouter LM.  Peripheral somatic nerve func-
population. Diabetes Care. 1993;16:1446–52. tion in relation to glucose tolerance in an elderly
54. Walters DP, Gatling W, Mullee MA, Hill RD.  The Caucasian population: the Hoorn study. Diabet Med.
prevalence of diabetic distal sensory neuropathy in 1996;13:960–6. https://doi.org/10.1002/(sici)1096-­
an English community. Diabet Med. 1992;9:349– 9136(199611)13:11<960::Aid-­dia268>3.0.Co;2-­z.
53. https://doi.org/10.1111/j.1464-­5491.1992. 66. Delcourt C, Vauzelle-Kervroedan F, Cathelineau G,
tb01795.x. Papoz L.  Low prevalence of long-term complica-
55. Jaiswal M, et al. Prevalence of and risk factors for tions in non-insulin-dependent diabetes mellitus in
diabetic peripheral neuropathy in youth with type 1 France: a multicenter study. CODIAB-INSERM-­
and type 2 diabetes: SEARCH for diabetes in youth ZENECA Pharma Study Group. J Diabetes
study. Diabetes Care. 2017;40:1226–32. https://doi. Complicat. 1998;12:88–95. https://doi.org/10.1016/
org/10.2337/dc17-­0179. s1056-­8727(97)98005-­3.
32 C. S. Hansen et al.

67. Janghorbani M, et  al. Peripheral neuropa- 79. Jensen TS, et al. A new definition of neuropathic pain.
thy in type 2 diabetes mellitus in Isfahan, Pain. 2011;152:2204–5. https://doi.org/10.1016/j.
Iran: prevalence and risk factors. Acta pain.2011.06.017.
Neurol Scand. 2006;114:384–91. https://doi. 80. Finnerup NB, et  al. Neuropathic pain: an updated
org/10.1111/j.1600-­0404.2006.00716.x. grading system for research and clinical practice.
68. Liu F, et al. Screening and prevalence of peripheral Pain. 2016;157:1599–606. https://doi.org/10.1097/j.
neuropathy in type 2 diabetic outpatients: a random- pain.0000000000000492.
ized multicentre survey in 12 city hospitals of China. 81. Galer BS, Gianas A, Jensen MP.  Painful diabetic
Diabetes Metab Res Rev. 2010;26:481–9. https:// polyneuropathy: epidemiology, pain descrip-
doi.org/10.1002/dmrr.1107. tion, and quality of life. Diabetes Res Clin Pract.
69. Gaede P, et  al. Multifactorial intervention and car- 2000;47:123–8.
diovascular disease in patients with type 2 diabe- 82. Davies M, Brophy S, Williams R, Taylor A.  The
tes. N Engl J Med. 2003;348:383–93. https://doi. prevalence, severity, and impact of painful diabetic
org/10.1056/NEJMoa021778. peripheral neuropathy in type 2 diabetes. Diabetes
70. Ismail-Beigi F, et  al. Effect of intensive treatment Care. 2006;29:1518–22. https://doi.org/10.2337/
of hyperglycaemia on microvascular outcomes in dc05-­2228.
type 2 diabetes: an analysis of the ACCORD ran- 83. Hébert HL, Veluchamy A, Torrance N, Smith
domised trial. Lancet. 2010;376:419–30. https://doi. BH.  Risk factors for neuropathic pain in diabe-
org/10.1016/S0140-­6736(10)60576-­4. tes mellitus. Pain. 2017;158:560–8. https://doi.
71. Su JB, et  al. HbA1c variability and diabetic org/10.1097/j.pain.0000000000000785.
peripheral neuropathy in type 2 diabetic patients. 84. Ponirakis G, et  al. Prevalence and risk factors for
Cardiovasc Diabetol. 2018;17:47. https://doi. painful diabetic neuropathy in secondary healthcare
org/10.1186/s12933-­018-­0693-­0. in Qatar. J Diabetes Investig. 2019;10:1558–64.
72. Fedele D, et  al. A multicenter study on the preva- https://doi.org/10.1111/jdi.13037.
lence of diabetic neuropathy in Italy. Diabetes 85. Ziegler D, et al. Painful and painless neuropathies are
Care. 1997;20:836–43. https://doi.org/10.2337/ distinct and largely undiagnosed entities in subjects
diacare.20.5.836. participating in an educational initiative (PROTECT
73. Ziegler D, et  al. Prevalence and clinical correlates study). Diabetes Res Clin Pract. 2018;139:147–54.
of cardiovascular autonomic and peripheral dia- https://doi.org/10.1016/j.diabres.2018.02.043.
betic neuropathy in patients attending diabetes cen- 86. Ziegler D, Rathmann W, Dickhaus T, Meisinger
ters. The Diacan Multicenter Study Group. Diabete C, Mielck A.  Neuropathic pain in diabetes, pre-
Metab. 1993;19:143–51. diabetes and normal glucose tolerance: the
74. Young MJ, Boulton AJM, Macleod AF, Williams MONICA/KORA Augsburg surveys S2 and
DRR, Sonksen PH.  A multicentre study of the S3. Pain Med. 2009;10:393–400. https://doi.
prevalence of diabetic peripheral neuropathy org/10.1111/j.1526-­4637.2008.00555.x.
in the United Kingdom hospital clinic popula- 87. Ziegler D, Rathmann W, Meisinger C, Dickhaus T,
tion. Diabetologia. 1993;36:150–4. https://doi. Mielck A. Prevalence and risk factors of neuropathic
org/10.1007/BF00400697. pain in survivors of myocardial infarction with
75. Kiani J, Moghimbeigi A, Azizkhani H, Kosarifard pre-diabetes and diabetes. The KORA Myocardial
S.  The prevalence and associated risk factors of Infarction Registry. Eur J Pain. 2009;13:582–7.
peripheral diabetic neuropathy in Hamedan, Iran. https://doi.org/10.1016/j.ejpain.2008.07.007.
Arch Iran Med. 2013;16:17–9. 88. Gylfadottir SS, et  al. Diabetic polyneuropathy
76. Cabezas-Cerrato J.  The prevalence of clinical dia- and pain, prevalence, and patient characteristics:
betic polyneuropathy in Spain: a study in primary a cross-sectional questionnaire study of 5,514
care and hospital clinic groups. Neuropathy Spanish patients with recently diagnosed type 2 diabetes.
Study Group of the Spanish Diabetes Society Pain. 2020;161:574–83. https://doi.org/10.1097/j.
(SDS). Diabetologia. 1998;41:1263–9. https://doi. pain.0000000000001744.
org/10.1007/s001250051063. 89. Gylfadottir SS, et  al. Diagnosis and prevalence of
77. Pai YW, Lin CH, Lee IT, Chang MH.  Prevalence diabetic polyneuropathy: a cross-sectional study
and biochemical risk factors of diabetic periph- of Danish patients with type 2 diabetes. Eur J
eral neuropathy with or without neuropathic pain Neurol. 2020;27:2575–85. https://doi.org/10.1111/
in Taiwanese adults with type 2 diabetes mellitus. ene.14469.
Diabetes Metab Syndr. 2018;12:111–6. https://doi. 90. Daousi C, et  al. Chronic painful peripheral neu-
org/10.1016/j.dsx.2017.09.013. ropathy in an urban community: a controlled
78. Kim SS, et al. Prevalence and clinical implications of comparison of people with and without diabe-
painful diabetic peripheral neuropathy in type 2 dia- tes. Diabet Med. 2004;21:976–82. https://doi.
betes: results from a nationwide hospital-based study org/10.1111/j.1464-­5491.2004.01271.x.
of diabetic neuropathy in Korea. Diabetes Res Clin 91. Abbott CA, Malik RA, van Ross ER, Kulkarni
Pract. 2014;103:522–9. https://doi.org/10.1016/j. J, Boulton AJ.  Prevalence and characteristics of
diabres.2013.12.003. painful diabetic neuropathy in a large community-­
The Epidemiology of Diabetic Neuropathy 33

based diabetic population in the U.K.  Diabetes investigation 2 diabetes (BARI 2D) Cohort. Diabetes
Care. 2011;34:2220–4. https://doi.org/10.2337/ Care. 2013;36:3208–15. https://doi.org/10.2337/
dc11-­1108. dc13-­0012.
92. Garoushi S, Johnson MI, Tashani OA.  A cross-­ 106. Sands ML, Shetterly SM, Franklin GM, Hamman
sectional study to estimate the point prevalence of RF.  Incidence of distal symmetric (sensory) neu-
painful diabetic neuropathy in Eastern Libya. BMC ropathy in NIDDM.  The San Luis Valley Diabetes
Public Health. 2019;19:78. https://doi.org/10.1186/ Study. Diabetes Care. 1997;20:322–9. https://doi.
s12889-­018-­6374-­9. org/10.2337/diacare.20.3.322.
93. Spallone V, Greco C.  Painful and painless dia- 107. Srinivasan S, et  al. Four-year Incident Neuropathy
betic neuropathy: one disease or two? Curr Diab and its Risk Factors in Subjects with Type 2 Diabetes.
Rep. 2013;13:533–49. https://doi.org/10.1007/ J Assoc Physicians India. 2019;67:34–7.
s11892-­013-­0387-­7. 108. Forrest KY, Maser RE, Pambianco G, Becker
94. Karlsson P, Hincker AM, Jensen TS, Freeman R, DJ, Orchard TJ.  Hypertension as a risk fac-
Haroutounian S. Structural, functional, and symptom tor for diabetic neuropathy: a prospective study.
relations in painful distal symmetric polyneuropa- Diabetes. 1997;46:665–70. https://doi.org/10.2337/
thies: a systematic review. Pain. 2019;160:286–97. diab.46.4.665.
https://doi.org/10.1097/j.pain.0000000000001381. 109. Pambianco G, et  al. The 30-year natural history
95. Daousi C, Benbow SJ, Woodward A, MacFarlane of type 1 diabetes complications: the Pittsburgh
IA.  The natural history of chronic painful periph- Epidemiology of Diabetes Complications Study
eral neuropathy in a community diabetes popula- experience. Diabetes. 2006;55:1463–9. https://doi.
tion. Diabet Med. 2006;23:1021–4. https://doi. org/10.2337/db05-­1423.
org/10.1111/j.1464-­5491.2006.01904.x. 110. Ou HT, Lee TY, Li CY, Wu JS, Sun ZJ.  Incidence
96. Benbow SJ, Chan AW, Bowsher D, MacFarlane IA, of diabetes-related complications in Chinese patients
Williams G.  A prospective study of painful symp- with type 1 diabetes: a population-based longitudinal
toms, small-fibre function and peripheral vascu- cohort study in Taiwan. BMJ Open. 2017;7:e015117.
lar disease in chronic painful diabetic neuropathy. https://doi.org/10.1136/bmjopen-­2016-­015117.
Diabet Med. 1994;11:17–21. 111. Papanas N, Ziegler D. Risk factors and comorbidities
97. Boulton AJ, Armstrong WD, Scarpello JH, Ward in diabetic neuropathy: an update 2015. Rev Diabet
JD. The natural history of painful diabetic neuropa- Stud. 2015;12:48–62. https://doi.org/10.1900/
thy–a 4-year study. Postgrad Med J. 1983;59:556–9. RDS.2015.12.48.
98. Ang L, Jaiswal M, Martin C, Pop-Busui R. Glucose 112. The Diabetes Control and Complications Trial
control and diabetic neuropathy: lessons from recent Research Group. The effect of intensive treatment
large clinical trials. Curr Diab Rep. 2014;14:528. of diabetes on the development and progression of
https://doi.org/10.1007/s11892-­014-­0528-­7. long-term complications in insulin-dependent dia-
99. Smith AG, et  al. Lifestyle intervention for pre-­ betes mellitus. N Engl J Med. 1993;329:977–86.
diabetic neuropathy. Diabetes Care. 2006;29:1294– https://doi.org/10.1056/nejm199309303291401.
9. https://doi.org/10.2337/dc06-­0224. 113. Ziegler D, Behler M, Schroers-Teuber M, Roden
100. Singleton JR, et  al. Exercise increases cutaneous M. Near-normoglycaemia and development of neu-
nerve density in diabetic patients without neuropa- ropathy: a 24-year prospective study from diagno-
thy. Ann Clin Transl Neurol. 2014;1:844–9. https:// sis of type 1 diabetes. BMJ Open. 2015;5:e006559.
doi.org/10.1002/acn3.125. https://doi.org/10.1136/bmjopen-­2014-­006559.
101. Malik RA, et  al. Sural nerve pathology in dia- 114. Patel A, et  al. Intensive blood glucose control and
betic patients with minimal but progressive neu- vascular outcomes in patients with type 2 diabe-
ropathy. Diabetologia. 2005;48:578–85. https://doi. tes. N Engl J Med. 2008;358:2560–72. https://doi.
org/10.1007/s00125-­004-­1663-­5. org/10.1056/NEJMoa0802987.
102. Pirart J. Diabetes mellitus and its degenerative com- 115. Callaghan BC, Little AA, Feldman EL, Hughes
plications: a prospective study of 4,400 patients RA.  Enhanced glucose control for prevent-
observed between 1947 and 1973 (author’s transl). ing and treating diabetic neuropathy. Cochrane
Diabete Metab. 1977;3:97–107. Database Syst Rev. 2012;6:CD007543. https://doi.
103. Pirart J. Diabetes mellitus and its degenerative com- org/10.1002/14651858.CD007543.pub2.
plications: a prospective study of 4,400 patients 116. Callaghan BC, et al. Association between metabolic
observed between 1947 and 1973 (2nd part) (author’s syndrome components and polyneuropathy in an
transl). Diabete Metab. 1977;3:173–82. obese population. JAMA Neurol. 2016;73:1468–76.
104. Pirart J. Diabetes mellitus and its degenerative com- https://doi.org/10.1001/jamaneurol.2016.3745.
plications: a prospective study of 4,400 patients 117. Markakis K, Bowling FL, Boulton AJ. The diabetic
observed between 1947 and 1973 (3rd and last part). foot in 2015: an overview. Diabetes Metab Res
Diabete Metab. 1977;3:245–56. Rev. 2016;32(1):169–78. https://doi.org/10.1002/
105. Pop-Busui R, et al. Impact of glycemic control strat- dmrr.2740.
egies ontheprogressionofdiabeticperipheral neu- 118. Brownrigg JR, et  al. The association of ulceration
ropathy in the bypass angioplasty revascularization of the foot with cardiovascular and all-cause mor-
34 C. S. Hansen et al.

tality in patients with diabetes: a meta-­ analysis. 132. Uhlig S, Meylan A, Rudolph U.  Reliability of
Diabetologia. 2012;55:2906–12. https://doi. short-term measurements of heart rate variability:
org/10.1007/s00125-­012-­2673-­3. findings from a longitudinal study. Biol Psychol.
119. Bjerg L, Hulman A, Charles M, Jørgensen ME, Witte 2020;2020:107905. https://doi.org/10.1016/j.
DR.  Clustering of microvascular complications in biopsycho.2020.107905.
Type 1 diabetes mellitus. J Diabetes Complicat. 133. Hojgaard MV, Holstein-Rathlou NH, Agner E,
2018;32:393–9. https://doi.org/10.1016/j. Kanters JK. Reproducibility of heart rate variability,
jdiacomp.2018.01.011. blood pressure variability and baroreceptor sensitiv-
120. Hsu WC, et  al. Somatic neuropathy is an inde- ity during rest and head-up tilt. Blood Press Monit.
pendent predictor of all- and diabetes-related 2005;10:19–24.
mortality in type 2 diabetic patients: a population-­ 134. Körei AE, et al. Why not to use the handgrip test in
based 5-year follow-up study (KCIS No. 29). the assessment of cardiovascular autonomic neu-
Eur J Neurol. 2012;19:1192–8. https://doi. ropathy among patients with diabetes mellitus? Curr
org/10.1111/j.1468-­1331.2011.03659.x. Vasc Pharmacol. 2017;15:66–73. https://doi.org/10.
121. Vinik AI, Ziegler D.  Diabetic cardiovas- 2174/1570161114666160822154351.
cular autonomic neuropathy. Circulation. 135. Bernardi L, et  al. Investigation methods for car-
2007;115:387–97. https://doi.org/10.1161/ diac autonomic function in human research stud-
CIRCULATIONAHA.106.634949. ies. Diabetes Metab Res Rev. 2011;27(7):654–64.
122. Spallone V, et  al. Recommendations for the use https://doi.org/10.1002/dmrr.1224.
of cardiovascular tests in diagnosing diabetic 136. von Scholten BJ, et  al. Cardiac autonomic func-
autonomic neuropathy. Nutr Metab Cardiovasc tion is associated with the coronary microcircu-
Dis. 2011;21:69–78. https://doi.org/10.1016/j. latory function in patients with type 2 diabetes.
numecd.2010.07.005. Diabetes. 2016;65:3129–38. https://doi.org/10.2337/
123. Ewing DJ, Clarke BF. Diabetic autonomic neuropa- db16-­0437.
thy: present insights and future prospects. Diabetes 137. Zobel EH, et  al. Cardiac autonomic function is
Care. 1986;9:648–65. associated with myocardial flow reserve in type 1
124. The Consensus Committee of the American diabetes. Diabetes. 2019;68:1277–86. https://doi.
Autonomic Society and the American Academy org/10.2337/db18-­1313.
of Neurology. Consensus statement on the defini- 138. Maser RE, et  al. Diabetic autonomic neuropathy
tion of orthostatic hypotension, pure autonomic and cardiovascular risk. Pittsburgh Epidemiology of
failure, and multiple system atrophy. Neurology. Diabetes Complications Study III. Arch Intern Med.
1996;46:1470. 1990;150:1218–22.
125. Papanas N, Ziegler D. Prediabetic neuropathy: does 139. Kempler P, et  al. Autonomic neuropathy is asso-
it exist? Curr Diab Rep. 2012;12:376–83. https://doi. ciated with increased cardiovascular risk fac-
org/10.1007/s11892-­012-­0278-­3. tors: the EURODIAB IDDM Complications
126. Diakakis GF, et al. Association of impaired glucose Study. Diabet Med. 2002;19:900–9. https://doi.
tolerance with increased heart rate and subclinical org/10.1046/j.1464-­5491.2002.00821.x.
inflammation. Hell J Cardiol. 2005;46:394–401. 140. Witte DR, et al. Risk factors for cardiac autonomic
127. Fiorentini A, Perciaccante A, Paris A, Serra P, neuropathy in type 1 diabetes mellitus. Diabetologia.
Tubani L.  Circadian rhythm of autonomic activ- 2005;48:164–71. https://doi.org/10.1007/
ity in non diabetic offsprings of type 2 diabetic s00125-­004-­1617-­y.
patients. Cardiovasc Diabetol. 2005;4:15. https:// 141. Tannus LR, Drummond KR, Clemente EL, da Matta
doi.org/10.1186/1475-­2840-­4-­15. Mde F, Gomes MB.  Predictors of cardiovascular
128. Gerritsen J, et al. Glucose tolerance and other deter- autonomic neuropathy in patients with type 1 dia-
minants of cardiovascular autonomic function: the betes. Front Endocrinol. 2014;5:191. https://doi.
Hoorn Study. Diabetologia. 2000;43:561–70. https:// org/10.3389/fendo.2014.00191.
doi.org/10.1007/s001250051344. 142. Pop-Busui R, et al. Effects of prior intensive insulin
129. Gerritsen J, et  al. Impaired autonomic function is therapy on cardiac autonomic nervous system func-
associated with increased mortality, especially in tion in type 1 diabetes mellitus: the Diabetes Control
subjects with diabetes, hypertension, or a history of and Complications Trial/Epidemiology of Diabetes
cardiovascular disease: the Hoorn Study. Diabetes Interventions and Complications study (DCCT/
Care. 2001;24:1793–8. EDIC). Circulation. 2009;119:2886–93. https://doi.
130. Singh JP, et  al. Association of hyperglycemia with org/10.1161/CIRCULATIONAHA.108.837369.
reduced heart rate variability (The Framingham 143. Metwalley KA, Hamed SA, Farghaly HS.  Cardiac
Heart Study). Am J Cardiol. 2000;86:309–12. autonomic function in children with type 1 diabe-
131. Stein PK, et al. The relationship of heart rate and heart tes. Eur J Pediatr. 2018;177:805–13. https://doi.
rate variability to non-diabetic fasting glucose levels org/10.1007/s00431-­018-­3122-­1.
and the metabolic syndrome: the Cardiovascular 144. Kim HY, Jung HW, Lee YA, Shin CH, Yang
Health Study. Diabet Med. 2007;24:855–63. https:// SW.  Cardiac autonomic neuropathy in nonobese
doi.org/10.1111/j.1464-­5491.2007.02163.x. young adults with type 1 diabetes. Ann Pediatr
The Epidemiology of Diabetic Neuropathy 35

Endocrinol Metabol. 2019;24:180–6. https://doi. 156. Mather KJ, et  al. Prevalence of microvascular and
org/10.6065/apem.2019.24.3.180. macrovascular disease in the glycemia reduction
145. Ratzmann KP, Raschke M, Gander I, Schimke E. approaches in diabetes  - a comparative effective-
Prevalence of peripheral and autonomic neuropathy ness (GRADE) study cohort. Diabetes Res Clin
in newly diagnosed type II (noninsulin-­dependent) Pract. 2020;165:108235. https://doi.org/10.1016/j.
diabetes. J Diabet Complications. 1991;5:1–5. diabres.2020.108235.
https://doi.org/10.1016/0891-­6632(91)90002-­7. 157. Valensi P, Pariès J, Attali JR.  Cardiac autonomic
146. Ziegler D, Dannehl K, Muhlen H, Spüler M, Gries neuropathy in diabetic patients: influence of diabetes
FA.  Prevalence of cardiovascular autonomic dys- duration, obesity, and microangiopathic complica-
function assessed by spectral analysis, vector anal- tions–the French multicenter study. Metabolism.
ysis, and standard tests of heart rate variation and 2003;52:815–20. https://doi.org/10.1016/
blood pressure responses at various stages of dia- s0026-­0495(03)00095-­7.
betic neuropathy. Diabet Med. 1992;9:806–14. 158. Low PA, et  al. Autonomic symptoms and diabetic
147. Jyotsna VP, Sahoo A, Sreenivas V, Deepak neuropathy: a population-based study. Diabetes
KK.  Prevalence and pattern of cardiac autonomic Care. 2004;27:2942–7.
dysfunction in newly detected type 2 diabetes mel- 159. Jaiswal M, et al. Cardiovascular autonomic neuropa-
litus. Diabetes Res Clin Pract. 2009;83:83–8. https:// thy in adolescents and young adults with type 1 and
doi.org/10.1016/j.diabres.2008.09.054. type 2 diabetes: The SEARCH for Diabetes in Youth
148. Arif ZA, Shaikh IA, Masood N.  Cardiovascular Cohort Study. Pediatr Diabetes. 2018;19:680–9.
autonomic neuropathy (CAN) in patients of type https://doi.org/10.1111/pedi.12633.
2 diabetes mellitus: a tertiary care hospital based 160. Migisha R, et  al. Prevalence and correlates of car-
study. Indian Heart J. 2014;66:751–4. https://doi. diovascular autonomic neuropathy among patients
org/10.1016/j.ihj.2014.10.417. with diabetes in Uganda: a hospital-based cross-­
149. Pop-Busui R, et  al. Effects of cardiac autonomic sectional study. Glob Heart. 2020;15:21. https://doi.
dysfunction on mortality risk in the action to con- org/10.5334/gh.765.
trol cardiovascular risk in diabetes (ACCORD) 161. Pan Q, et  al. Prevalence and diagnosis of dia-
trial. Diabetes Care. 2010;33:1578–84. https://doi. betic cardiovascular autonomic neuropathy in
org/10.2337/dc10-­0125. Beijing, China: a retrospective multicenter clinical
150. Zoppini G, et  al. Prevalence of cardiovascular study. Front Neurosci. 2019;13:1144. https://doi.
autonomic neuropathy in a cohort of patients with org/10.3389/fnins.2019.01144.
newly diagnosed type 2 diabetes: the verona newly 162. Tang M, Donaghue KC, Cho YH, Craig
diagnosed type 2 diabetes study (VNDS). Diabetes ME.  Autonomic neuropathy in young people
Care. 2015;38:1487–93. https://doi.org/10.2337/ with type 1 diabetes: a systematic review. Pediatr
dc15-­0081. Diabetes. 2013;14:239–48. https://doi.org/10.1111/
151. Hansen CS, et al. Vitamin B12 deficiency is associ- pedi.12039.
ated with cardiovascular autonomic neuropathy in 163. Veglio M, et  al. Autonomic neuropathy in non-­
patients with type 2 diabetes. J Diabetes Complicat. insulin-­dependent diabetic patients: correlation with
2016;31(1):202–8. https://doi.org/10.1016/j. age, sex, duration and metabolic control of diabetes.
jdiacomp.2016.08.025. Diabete Metab. 1990;16:200–6.
152. Chung JO, et  al. Serum apolipoprotein A-1 con- 164. Gæde P, et  al. Years of life gained by multifacto-
centrations and the prevalence of cardiovascular rial intervention in patients with type 2 diabetes
autonomic neuropathy in individuals with type 2 mellitus and microalbuminuria: 21 years follow-
diabetes. J Diabetes Complicat. 2018;32:357–61. ­up on the Steno-2 randomised trial. Diabetologia.
https://doi.org/10.1016/j.jdiacomp.2018.01.006. 2016;59:2298–307. https://doi.org/10.1007/
153. Andersen ST, et al. Risk factors for the presence and s00125-­016-­4065-­6.
progression of cardiovascular autonomic neuropathy 165. Reichard P.  Risk factors for progression of
in type 2 diabetes: ADDITION-Denmark. Diabetes microvascular complications in the Stockholm
Care. 2018;41:2586–94. https://doi.org/10.2337/ Diabetes Intervention Study (SDIS). Diabetes
dc18-­1411. Res Clin Pract. 1992;16:151–6. https://doi.
154. Ko SH, et  al. Cardiovascular autonomic dysfunc- org/10.1016/0168-­8227(92)90087-­8.
tion predicts acute ischaemic stroke in patients 166. Tang Y, et  al. Intensive risk factor manage-
with Type 2 diabetes mellitus: a 7-year follow-up ment and cardiovascular autonomic neuropathy
study. Diabet Med. 2008;25:1171–7. https://doi. in type 2 diabetes: the ACCORD trial. Diabetes
org/10.1111/j.1464-­5491.2008.02567.x. Care. 2021;44:164–73. https://doi.org/10.2337/
155. Bhuyan AK, Baro A, Sarma D, Choudhury B.  A dc20-­1842.
study of cardiac autonomic neuropathy in patients 167. Choung RS, et al. Risk of gastroparesis in subjects
with type 2 diabetes mellitus: a Northeast India expe- with type 1 and 2 diabetes in the general population.
rience. Indian J Endocrinol Metabol. 2019;23:246– Am J Gastroenterol. 2012;107:82–8. https://doi.
50. https://doi.org/10.4103/ijem.IJEM_336_18. org/10.1038/ajg.2011.310.
36 C. S. Hansen et al.

168. Horowitz M, et al. Relationships between oesopha- 172. Albers JW, Pop-Busui R.  Diabetic neuropathy:
geal transit and solid and liquid gastric emptying in mechanisms, emerging treatments, and subtypes.
diabetes mellitus. Eur J Nucl Med. 1991;18:229–34. Curr Neurol Neurosci Rep. 2014;14:473. https://doi.
https://doi.org/10.1007/bf00186645. org/10.1007/s11910-­014-­0473-­5.
169. Parkman HP, Hasler WL, Fisher RS.  American 173. Albers JW, Brown MB, Sima AA, Greene
Gastroenterological Association technical review DA.  Frequency of median mononeuropathy in
on the diagnosis and treatment of gastroparesis. patients with mild diabetic neuropathy in the early
Gastroenterology. 2004;127:1592–622. https://doi. diabetes intervention trial (EDIT). Tolrestat Study
org/10.1053/j.gastro.2004.09.055. Group For Edit (Early Diabetes Intervention Trial).
170. Camilleri M.  Clinical practice. Diabetic gastropa- Muscle Nerve. 1996;19:140–6.
resis. N Engl J Med. 2007;356:820–9. https://doi. 174. Chan YC, Lo YL, Chan ES.  Immunotherapy
org/10.1056/NEJMcp062614. for diabetic amyotrophy. Cochrane Database
171. Kempler P, et  al. Management strategies for gas- Syst Rev. 2017;7:Cd006521. https://doi.
trointestinal, erectile, bladder, and sudomotor dys- org/10.1002/14651858.CD006521.pub4.
function in patients with diabetes. Diabetes Metab 175. Gibbons CH.  Treatment-induced neuropathy of
Res Rev. 2011;27:665–77. https://doi.org/10.1002/ diabetes. Curr Diab Rep. 2017;17:127. https://doi.
dmrr.1223. org/10.1007/s11892-­017-­0960-­6.
Clinical Features of Diabetes
Neuropathies

Gordon Sloan, Qi Pan, Ling Gao, Lixin Guo,


and Solomon Tesfaye

1 Introduction limb amputation in diabetes. However, a number


of other syndromes may also present in diabetes
Patients with diabetes may present with a wide which involve the cranial nerves, peripheral
range of neuropathic syndromes with varied clin- nerves, radicular nerves, nerve plexus, the and
ical presentations, disease aetiologies, prognosis, autonomic nervous system. This chapter will
and treatments. By far the commonest and most describe the classification of diabetic neuropa-
well understood is diabetic peripheral neuropathy thies and their clinical features, with the main
(DPN). DPN is a chronic and progressive condi- emphasis on neuropathies of the lower
tion, defined as ‘a symmetrical, length-dependent extremity.
sensorimotor polyneuropathy attributable to met-
abolic and microvessel alterations as a result of
chronic hyperglycaemia exposure (diabetes) and 2 Epidemiology
cardiovascular risk covariates’ [1]. This condi-
tion is the precipitant for two of the most trouble- There is a great variation in the reported preva-
some chronic complications of diabetes, lence of DPN. This is likely secondary to differ-
painful-DPN, and diabetic foot diseases (i.e., foot ing study methods; although regional/population
ulceration, Charcot neuroarthropathy, and infec- variation may also contribute. The overall preva-
tion). The latter is the main precipitant for lower lence of clinical DPN is estimated at approxi-
mately 30% [2]. Where sensitive diagnostic
techniques are used, such as nerve conduction
studies, the prevalence rises to over 50%. The
G. Sloan prevalence of DPN is dependent upon the dura-
Diabetes Research Unit, Sheffield Teaching Hospitals
Trust and the University of Sheffield, Sheffield, UK tion of diabetes. In type 1 diabetes mellitus
(T1DM), newly diagnosed patients have a low
Q. Pan · L. Guo
Department of Endocrinology, National Center of prevalence of DPN, but in newly diagnosed type
Gerontology, Beijing Hospital, Beijing, China 2 diabetes mellitus (T2DM) the prevalence is
L. Gao approximately 13% [3]. The most important risk
Department of Endocrinology and Metabolism, factor for DPN is HbA1c followed by duration of
Renmin Hospital of Wuhan University, Wuhan, China diabetes/age [2]. However, other modifiable car-
S. Tesfaye (*) diovascular risk factors have also been shown to
Diabetes Research Unit, Sheffield Teaching Hospitals be independent predictors of DPN [4].
and the University of Sheffield, Sheffield, UK
e-mail: solomon.tesfaye@nhs.net

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 37


S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_3
38 G. Sloan et al.

The prevalence of autonomic neuropathy in plexus neuropathy to be higher in diabetes


diabetes is also heavily dependent upon the study (2.79/100,000/year [95% CI 1.94–3.64]) com-
methods, particularly the diagnostic technique pared to non-diabetes patients (1.27/100,000/
and case definition. The prevalence of cardiovas- year [95% CI 0.71–1.83]) [9]. Chronic inflamma-
cular autonomic neuropathy may be at least 30% tory demyelinating polyneuropathy has classi-
in patients with type 1 diabetes after 20 years dis- cally been described to be more common in
ease duration and 60% in patients with type 2 diabetes, although epidemiological studies have
diabetes after 15 years [5]. The epidemiological failed to show an association [10].
data on other diabetic autonomic neuropathies is
limited, although the cumulative incidence of
diabetic gastroparesis over 10 years was higher in 3 Classification
type 1 diabetes (5%) than in type 2 diabetes (1%)
and in control subjects (1%). The diabetic neuropathies have a heterogeneous
The prevalence of other diabetic neuropathies clinical presentation, natural history, pattern of
is even less well known. Median and ulnar nerve neurological impairment, underlying nerve
entrapment may occur in over 30% of patients pathology, risk factors, and disease mechanisms
with type 1 or 2 diabetes [6, 7]. In a retrospective (Fig.  1). A number of different classification
study of 8150 patients with diabetes who were schemes for diabetic neuropathies have been pro-
hospitalized in Marsala, Italy, over a 12-year posed over the years. The most recent and widely
period, 0.75% had evidence of cranial nerve pal- used classification scheme in the Position
sies, with third nerve palsies the most common Statement by the American Diabetes Association
(0.35%), followed by seventh (0.21%) and sixth stratifies syndromes according to their neurologi-
(0.15%) nerve palsies [8]. The prevalence of cra- cal pattern of involvement (Table 1). All of these
nial nerve palsies in the community, however, is neuropathic syndromes may present in patients
unknown. A study in Olmsted County, Minnesota without diabetes with other underlying disease
found the prevalence of lumbosacral radiculo- mechanisms. Moreover, patients with diabetes

Polyneuropathy Mononeuropathies Plexopathy Autonomic neuropathy

Fig. 1  Clinical presentation of different neuropathic syndromes in diabetes (reproduced from [11])
Clinical Features of Diabetes Neuropathies 39

Table 1  Classification of diabetic neuropathies (adapted are more likely to experience entrapment
from Pop-Busui et al. [5])
neuropathies.
Classification of diabetic neuropathies
Generalized symmetric polyneuropathies
 • Diabetic sensorimotor polyneuropathy
   – Mixed sensorimotor neuropathy
4 Clinical Features of Diabetic
   – Predominantly large fibre neuropathy Neuropathies
   – Predominantly small fibre neuropathy
   – Pure small fibre neuropathy 4.1 Diabetic Peripheral
 • Acute painful-distal sensorimotor
polyneuropathies
Neuropathy
   – Treatment induced neuropathy of diabetes
[TIND] In DPN there is a ‘length-related’ pattern of neu-
   – Hyperglycaemia induced ronal dysfunction, with sensory symptoms/signs
 • Autonomic neuropathy starting symmetrically in the toes of both feet and
   – Cardiovascular autonomic neuropathy
    Reduced heart rate variability then extending as the disease advances to involve
    Resting tachycardia the rest of the foot and lower limb in a ‘stocking’
    Orthostatic hypotension distribution (Fig. 2). Once the disease has reached
    Sudden cardiac death approximately the level of the knees it usually is
    Exercise intolerance
    Silent myocardial ischaemia also present in the distal upper-limb, the finger-
   – Gastrointestinal autonomic neuropathy tips. With upper limb involvement, there is a
    Oesophageal dysmotility similar symmetrical pattern of progression proxi-
    Gastroparesis mally in a ‘glove’ distribution. Although the
    Diabetic diarrhoea
    Constipation nerve damage can extend over the entire body
    Faecal incontinence including the trunk, and even the head and face,
   – Urogenital this is exceptional. As the disease advances, overt
    Neurogenic bladder motor manifestations such as wasting of the
    Sexual dysfunction (erectile and female sexual
dysfunction; retrograde ejaculation) small muscles of the hands and limb weakness
   – Sudomotor dysfunction become apparent. The precise temporal course of
    Anhidrosis nerve fibre injury is unclear, it has been proposed
    Hyperhidrosis that sensory small-fibre dysfunction occurs first,
    Gustatory sweating
   – Metabolic (i.e., hypoglycaemia unawareness) followed by sensory and motor large-fibre; how-
   – Pupillary dysfunction ever, there is no clear evidence for this hypothesis
Focal/multifocal neuropathies [11]. Isolated small-fibre neuropathy is a possible
 • Mononeuropathy manifestation of DPN, although this is uncom-
   – Mononeuropathies mon (approximately 2.5%) [12]. When present,
    Cranial nerve [e.g., oculomotor nerve palsy,
facial nerve palsy] pure small-fibre neuropathy involves clinical loss
    Peripheral nerve [e.g., peroneal nerve palsy] of modalities serving pain and thermal sensation
 • Mononeuritis multiplex but preservation of large fibre modalities (such as
 • Radiculoplexus neuropathy vibration, light touch, proprioception, and motor
   – Radiculopathy
function).
    Thoracic
    Abdominal The clinical presentation of DPN is often
    Thoraco-abdominal insidious. Patients may be entirely asymptom-
   – Plexopathy atic; hence the critical importance of diabetic
    Lumbosacral plexopathy
foot screening, as all too often the first presenta-
    Cervical plexopathy
 • Entrapment neuropathies (e.g. carpal tunnel tion of DPN is with a diabetic foot ulcer. The sen-
syndrome, ulnar neuropathy) sory symptoms which develop in DPN are often
divided into ‘positive’ and ‘negative’ symptoms,
the former referring to paresthesias/painful neu-
ropathic symptoms and the latter to symptoms of
40 G. Sloan et al.

Slight sensory loss Moderate sensory loss Extensive sensory loss


Diminished/absent sensation in Significantly impaired/absent Absent sensation in the foot and
toes (light touch/vibration/pin- sensation in the foot extending up impaired sensation in the upper
prick) the leg – risk of foot ulceration and lower limb (rarely the trunk)
Normal motor examination Absent ankle reflex Weakness and/or wasting in the
except ankle reflex may be Foot deformity lower/upper limb
diminished/absent Weakness in toe/ankle Impaired knee reflex
flexion/extension Gait/balance impairment

Fig. 2  Clinical presentation and progression of sensory changes in diabetic peripheral neuropathy (reproduced from
[11])

sensory loss. Negative symptoms may be sensory abnormalities. Innervation of sweat


described as an awareness of abnormal sensation glands is also impaired resulting in ‘sudomotor
such as a ‘numbness’ or ‘deadness’ in their feet. dysfunction’, which leads to skin dryness and
Patients may describe feeling as if their socks are cracking, acting as a potential precipitant for
always bunched up in their shoes or as if they are infection and foot ulceration. Early motor mani-
walking on pebbles or cotton wool. Whereas pos- festations include the absence or reduction in
itive symptoms may be non-painful, such as tin- ankle reflexes bilaterally. Reduce ankle reflexes
gling, pressing, tightness or pins and needles, or may be present in elderly patients without
painful, as in painful-DPN.  Sensory symptoms DPN. Another motor sign is foot deformity, such
progress in the aforementioned ‘stocking/glove as clawing of the toes and prominent metatarsal
distribution’, although different regions of the heads as a result of denervation of the interphalan-
body may experience different symptoms. For geal extensor and metacarpal-phalangeal flexor
example, in advanced disease patients may expe- muscles. More advanced clinical motor findings
rience neuropathic pain in the upper limb whereas include motor weakness, wasting of the small
the lower-limb may have only negative symp- muscles of the foot and extensor hallucis longus,
toms. Patients may also notice impaired balance high arching of the foot and the absence of knee
and gait with advanced lower limb neuropathy. reflexes. The presence of foot deformity and sen-
Once the disease affects the hands patients may sory loss in the foot predispose the patient to com-
initially feel reduced sensation of finger-prick plications such as foot ulceration and Charcot
blood glucose testing before developing pro- neuroarthropathy. Similar to foot involvement, the
found difficulties with day-to-day tasks such as distal extremity of the limb, the tips of the fingers,
typing, opening tins/jars, and gripping. are affected first before spreading proximally in a
The early clinical signs of DPN include a distal ‘glove’ distribution. As upper limb involvement
loss of light touch, temperature, and pin-prick progresses sensory loss becomes more advanced
sensation before proprioception and vibration and there is weakness with wasting of the muscles
Clinical Features of Diabetes Neuropathies 41

of the hands. However, advanced upper limb descriptors of pain are burning (>60%) and elec-
involvement in DPN is rare, and other causative tric/shocks (40–50%) [12, 17, 18]. The preva-
conditions should be considered, such as entrap- lence of other symptoms is less clear although
ment neuropathy or spinal cord pathology. sharp, aching, and cold pains may also be pres-
To complete the examination of the foot it is ent. The pain is often very distressing and more
important to examine the skin, joints, and vascu- than half of those with painful-DPN grade their
lar supply of the foot and the footwear of the symptoms as severe [19]. The pain may occur
patient. The neuropathic foot may be warm with intermittently throughout the day, but more com-
a bounding arterial pulse, due to peripheral auto- monly it is constant with nocturnal exacerbation,
nomic neuropathy. However, peripheral vascular leading to disruption of sleep in up to three quar-
disease is a common co-morbidity in DPN. This ters of individuals [20]. Similar to painless-DPN
may manifest itself as peripheral hair loss, cool/ the course of the disease over time has not been
cold skin, pallor and reduced/absent peripheral thoroughly investigated. A questionnaire based
pulses. A thorough assessment of the patients’ study (n = 105) found that in 72% of patients the
footwear is also mandatory. Patients with DPN disease had worsened since onset, 12% reported
commonly wear shoes which are too small as the pain improved and in 15% it had stayed the
they ‘feel comfortable’. Furthermore, it is possi- same [17]. Furthermore, longitudinal studies are
ble to detect areas of wear from within the shoe. necessary to examine the natural history of
Trauma resulting from a poor fit, abnormal wear painful-DPN.
from internal pressure areas and foreign objects
are common contributors to the pathway to foot 4.1.2 Co-morbidities of Diabetic
ulceration [13]. Peripheral Neuropathy
It is important to consider and address other
4.1.1 Painful Diabetic Peripheral sequalae of diabetes and DPN and related con-
Neuropathy ditions as part of the holistic assessment of the
Despite patients with painful-DPN often suffering patient with DPN. Most pressingly, DPN is the
with severe neuropathic pain. There are generally main precipitant for diabetic foot disease such
few clinical signs or neurological features to dif- as foot ulceration and Charcot Neuroarthropathy,
ferentiate these cases from painless-DPN [14]. It which can lead to lower limb amputations.
has been reported that the neuropathic deficit in Co-morbid vascular disease, including cardio-
painful-DPN may be more advanced, but the clin- vascular disease, medial arterial calcification,
ical examination is normally indistinct from pain- and peripheral vascular disease, is also common
ful- and painless-DPN.  Patients may report in patients with DPN [21]. Moreover, there is an
hyperalgesia (increased pain from a stimulus that association between DPN, foot ulceration, and
normally provokes pain) and allodynia (pain from amputation and a high risk of death [22]. The
an innocuous stimuli), although clinically repro- other microvascular complications of diabetes,
ducible brush-evoked allodynia is rare [15]. such a autonomic neuropathy, nephropathy, and
The symptoms of painful-DPN widely vary retinopathy, may also be present and there are
from patient to patient. It can occur at any sever- also potential associations with DPN and
ity of neuropathic deficit in DPN. Indeed, severe obstructive sleep apnoea and cognitive dysfunc-
sensory loss in the presence of painful neuro- tion [21].
pathic symptoms might occur, the so-called pain- Unsurprisingly, patients with DPN can
ful/painless leg [16]. The distribution of the pain develop mood disorders, such as depression, and
depends upon the severity of neuronal loss, in can have a reduced quality of life [21]. Moreover,
mild cases the pain may be present in only the the neuropathic symptoms in painful-DPN can
toes; whereas advanced cases may have upper cast a huge burden on all aspects of patients’
limb neuropathic pain. The most common lives, resulting in a limitation in all activities of
42 G. Sloan et al.

daily living, including their employment [12]. Such atypical features might include: weight
Therefore, it is common that these patients have loss; an absence of other complications of diabe-
depression, anxiety, sleep impairment and a pro- tes; asymmetry; rapidly progressing symptoms;
found reduced quality of life. predominant motor signs; predominant upper
limb signs; and evidence of multi-system
4.1.3 Differential Diagnosis ­disorders (i.e., malignancy, autoimmune disease,
of Diabetic Peripheral connective tissue disease, vasculitis, etc.). If a
Neuropathy diagnosis of a peripheral neuropathy/polyneu-
The potential differential diagnosis of DPN is ropathy is made, a careful clinical history and
vast. If there are clinical features present which examination and simple investigations (e.g., thy-
are not typical for DPN, then alternative diagno- roid function tests, vitamin B12, serum/urine
ses should be considered and excluded (Table 2). electrophoresis, full blood count and inflamma-
tory markers) is usually sufficient to exclude
other neuropathies.
Table 2  Differential diagnosis of DPN
Differential diagnosis of DPN
 • Metabolic 4.2 Acute Painful-Distal
   – Hypothyroidism
   – Vitamin deficiency (B12, thiamine, vitamin E,
Sensorimotor
post-gastric surgery, B6, pyridoxine, etc.) Polyneuropathies
   – Uraemia
   – Copper deficiency Acute painful neuropathies are rare transient neu-
   – Liver cirrhosis
   – Critical illness
ropathic syndromes presenting in a peripheral dis-
   – Amyloidosis tribution, which develop over a relatively short
   – Porphyria period of time, i.e. days to weeks. Sensory and
 • Drugs and toxins motor neuropathic deficits are often mild, but neu-
   – Alcohol ropathic pain may be severe. There are two distinct
   – Cytotoxic drugs (e.g., vinca alkaloids, platinum
based agents, bortezomib and thalidomide, etc. syndromes, the first of which occurs within the
   – Antibiotics (e.g., chlorambucil, nitrofurantoin, context of poor glycaemic control, and the second
isoniazid, linezolid, metronidazole) with rapid improvement in glycaemic control
   – Anti-retroviral agents (Treatment induced neuropathy of diabetes; TIND).
   – Immunosuppressant agents (e.g., interferons
and monoclonal antibodies)
 • Infectious 4.2.1 Hyperglycaemia Induced Acute
   – HIV Painful Distal Sensorimotor
   – Leprosy Polyneuropathy
   – Guillain-Barre syndrome
This condition is rare and occurs in patients with
   – Lyme borreliosis
   – Viral hepatitis diabetes and poor glycaemic control. It is poorly
   – Neurosyphilis understood as with improvements in the diagno-
 • Inherited sis and treatment of diabetes it is rarely seen in
   – Charcot-Marie-Tooth disease developed countries. The condition is associated
   – Hereditary sensory neuropathies
   – Fabry disease with burning pain and allodynia, which is worse
   – Other rare inherited neuropathies at night. The condition is also associated with
 • Inflammatory concomitant severe weight loss. There is gener-
   – Chronic inflammatory demyelinating ally a complete resolution of symptoms within 12
polyneuropathy
months, and weight gain with improvement in
   – Sarcoidosis
   – Autoimmune disorders (e.g., connective tissue glycaemic control.
diseases)
   – Vasculitis (e.g., polyarteritis nodosa)
 • Paraneoplastic neuropathy
Clinical Features of Diabetes Neuropathies 43

4.2.2 Treatment Induced Neuropathy Moreover, there may be an association with


of Diabetes TIND and prior weight loss/diabetic anorexia
Although TIND is better understood than hyper- [23, 25]; although, TIND itself is not believed to
glycaemia induced acute painful distal sensorim- present with weight loss unlike some other dia-
otor polyneuropathy there is still limited data betic neuropathies (e.g., hyperglycaemia induced
relating to the condition. This condition is also acute painful distal sensorimotor polyneuropathy
known as ‘insulin neuritis’, although this is a and radiculoplexus neuropathies).
misnomer as the condition is unrelated to the The symptoms of TIND develop approxi-
therapeutic agent used but rather the speed in mately 2–6 weeks following the improvement in
which glucose is lowered. A retrospective study glucose control [23]. The most common symp-
found that a 2% reduction in HbA1c over 3 toms include burning and shooting pain, and par-
months was associated with an approximate 10% aesthesia with hyperalgesia and allodynia in the
risk of TIND; whereas a 5% reduction is associ- distribution of the neuropathy. The symptoms are
ated with an over 90% risk of developing the con- in a peripheral neuropathy distribution, similar to
dition [23]. A greater degree of HbA1c reduction DPN. With mild reductions in HbA1c, the symp-
was shown to cause greater severity and more toms may only involve the toes and feet; how-
widespread distribution of neuropathy (Fig.  3). ever, with greater reduction in HbA1c the upper
Therefore, the condition is readily identifiable in limbs and even abdomen and trunk can be
the clinical context of neuropathic symptoms in involved in severe cases. Similar to painful-DPN,
the presence of improved glucose control. the pain is associated with disturbances in sleep.

Decrease in HbA1C
2-3 points 3-5 points 5+ points
Risk of TIND Development

Greater Decrease in HbA1C

Fig. 3  Clinical phenotype in treatment induced neuropathy of diabetes (reproduced from [24]
44 G. Sloan et al.

The examination findings are often mild but may 4.2.4 Cardiovascular Autonomic
involve sensory loss (reduced/absent pinprick Neuropathy
and/or vibration sensation at the toe/foot) and The presence of CAN holds clinical relevance as
distal toe extensor/flexor weakness and ankle it confers an increased risk of silent ischaemia,
reflex impairment. Neurophysiological findings cardiovascular events, arrhythmias, and cardio-
may reveal mild sensory, axonal neuropathy, and vascular mortality [5, 26]. The condition is read-
intra-epidermal nerve fibre density on skin biopsy ily identifiable in patients with longstanding
may be reduced [23, 25]. The condition is also diabetes, particularly those with DPN with which
frequently associated with autonomic symptoms, it shares risk factors, using sensitive autonomic
most commonly orthostatic intolerance and syn- function tests. However, overt clinical manifesta-
cope/pre-syncope although gastrointestinal tions are less common, but can be disabling.
symptoms and sweating alterations may occur Symptoms may include intolerance of exercise,
with greater reductions in HbA1c [23]. Finally, i.e. breathlessness on exertion, due to impaired
the condition is also associated with an increased heart rate variability, and resting tachycardia.
incidence of other microvascular complications Orthostatic symptoms suggestive of a postural
of diabetes, retinopathy, and nephropathy. drop in blood pressure (>20 mmHg) are gener-
There is scant evidence for the natural historyally associated with advanced disease.
of TIND.  It is classically described to improve Participants may experience light-headedness,
over a period of 12–18 months. A publication pre-syncope or syncope on standing, or some-
assessed 26 individuals with T1DM longitudi- times from lying to sitting. Anyone with sus-
nally over 8 years with comprehensive neurologi- pected orthostatic hypotension due to CAN
cal and clinical testing [25]. Patients were should undergo a careful review of their medica-
grouped into ‘stable glycaemic control’ and tions. Many drugs prescribed in patients with
‘unstable glycaemic control’, the latter of whom advanced diabetes or DPN may cause or worsen
developed a further episode of TIND.  Patients orthostatic hypotension, e.g. antihypertensives,
with stable glycaemic control had improvements SGLT-2 inhibitors, diuretics, and anticholinergic
in intra-epidermal nerve fibre density, neurologi- agents such as tricyclic antidepressants. Cardiac
cal examination and nerve conduction measures ischaemia/infarction may also present atypically
after 8 years; whereas the unstable group had a in patients with CAN due to ‘silent ischaemia’.
deterioration in these parameters. Therefore, ischaemia/infarction may be missed
entirely or present atypically. In the acutely
4.2.3 Autonomic Neuropathy unwell patient with diabetes silent myocardial
Diabetic autonomic neuropathies are sub-­ infarction should be considered and investiga-
classified according to the organ or body system tions such as electrocardiograms or serum tropo-
affected. Abnormalities in autonomic function nin levels may need to be performed to exclude
tests are common in patients with longstanding the condition.
diabetes; however, clinically significant auto-
nomic symptoms are uncommon. Other than spe- 4.2.5 Gastrointestinal Autonomic
cific scenarios, such as TIND, the condition Neuropathy
generally has a gradual onset and is slowly pro- Patients with diabetes commonly experience
gressive. Cardiac autonomic neuropathy is troublesome gastrointestinal symptoms which
believed to be the most common; however, the can be severe, causing great patient morbidity.
epidemiological data is heavily dependent upon These symptoms can be due to autonomic neu-
the studied population, diagnostic criteria, and ropathy, but other factors should be considered
investigative method. Autonomic neuropathies such as autoimmune disorders (e.g., coeliac dis-
are discussed in more detail in Chaps. 12, 26, 27, ease and hypo/hyperthyroidism), medications
and 28. (e.g., metformin and GLP-1 agonists), gastroduo-
denal ulceration, and hyperglycaemia. Autonomic
Clinical Features of Diabetes Neuropathies 45

neuropathy in diabetes can affect any part of the Table 3  Differentiation of entrapment and microvascu-
litic causes of mononeuropathies in diabetes
gastrointestinal tract. The most well described
and common presentation in the upper gastroin- Microvasculitic Entrapment
mononeuropathy neuropathy
testinal tract is gastroparesis. Gastroparesis leads
Onset Sudden Gradual
to a slower transit of food through the stomach
Natural Monophasic, Progressive
which may present with nausea, vomiting, and history maximal deficit at deterioration in
abdominal bloating. Moreover, there may be glu- onset of symptoms symptoms
cose variability with unexplained hypoglycaemia with spontaneous without treatment
resolution
due to dissociation between food absorption and
Location Cranial nerves: CN Upper limb:
insulin administration [5]. Other upper gastroin- III, IV, VI, VII, and median and ulnar
testinal symptoms of autonomic neuropathy may others more rarely nerve
include dysphagia and heart burn. Lower gastro- Peripheral nerves: Lower limb:
intestinal symptoms range from constipation to any nerve; peroneal, peroneal, femoral,
femoral, radial, lateral cutaneous
diarrhoea and faecal incontinence. Indeed, diar- ulnar, and median nerve of the thigh,
rhoea may alternate with constipation. Diabetic more common lateral plantar
diarrhoea is often worse at night and may be nerve
associated with small bowel bacterial Symptoms Acute onset of pain Paraesthesia and
and neurological pain within the
overgrowth.
dysfunction in the territory of the
territory of the nerve with motor
4.2.6 Urogenital Autonomic nerve signs occurring
Neuropathy late
Urinary autonomic dysfunction may lead to hesi-
tancy or increased frequency of micturition. In
serious cases there is urinary retention associated 4.3 Mononeuropathies
with overflow incontinence. Women with diabe-
tes may have reduced sexual desire and increased Diabetic mononeuropathies may occur second-
pain during intercourse due to inadequate vaginal ary to compression (entrapment neuropathies) or
lubrication [27]. Erectile dysfunction is common microvasculitis. It is important to distinguish
in men with diabetes; however, its aetiology is between the two as the former can respond well
often multi-faceted, other contributory factors to treatment, such as surgical release of the nerve
including vascular disease, metabolic control, (Table  3). Moreover, other causes should be
nutrition, endocrine disorders (e.g., hypogonad- actively sought and excluded as the differential
ism), psychogenic factors, and medications [27]. diagnosis of mononeuropathies includes serious
causes such cerebrovascular disease.
4.2.7 Sudomotor Dysfunction
Sudomotor dysfunction can lead to anhidrosis or 4.3.1 Microvasculitic
hyperhidrosis. Anhidrosis is relevant to the Mononeuropathies
patient with DPN where peripheral anhidrosis Microvasculitic diabetic mononeuropathies gen-
can lead to drying and cracking of the skin, which erally may involve the cranial nerves or periph-
can precipitate bacterial infection of the foot. eral nerves. The microvasculitic neuropathies
Hyperhidrosis can be distressing and embarrass- present with a rapid onset nerve palsy, often asso-
ing to patients. It is often associated with eating; ciated with pain [28]. The specific symptoms
in this situation it is known as gustatory depend upon the affected nerve. The most com-
sweating. mon cranial nerve involved is the third. Patients
with this condition present with sudden onset
orbital/frontal pain associated with ptosis and
ophthalmoplegia with pupillary sparing. Fourth
and sixth nerve neuropathies would be associated
46 G. Sloan et al.

with ophthalmoplegia and facial nerve palsy distribution of the nerve. Sensory signs such as a
occurs with seventh nerve involvement. The dif- reduction in sensation in the territory of the
ferential diagnosis of cranial nerve palsy is vast nerve can be detected. Motor signs such as weak-
and should not be attributed to diabetes without a ness and muscle wasting of the innervated mus-
careful clinical history, examination, investiga- cles are late signs. Median and ulnar nerve
tions, and specialist input. Cerebral imaging with entrapment neuropathies may occur concur-
computer tomography or magnetic resonance rently and can be bilateral. However, if this is the
imaging (MRI) is generally required to exclude case other causes should be considered and the
structural compression of the nerve in question or patient referred for careful neurophysiological
cerebrovascular disease. testing. Indeed, the diagnosis of entrapment neu-
Peripheral mononeuropathies may involve ropathies often requires nerve conduction stud-
any nerve, including the peroneal, sural, sciatic, ies; however, this is particularly challenging if
femoral, ulnar, and median nerve [28]. Similar to there is concurrent DPN. Without treatment the
cranial nerve involvement the presentation is sud- nerve dysfunction is typically progressive and
den. There may be motor and/or sensory signs in can lead to irreversible neurological impairment
the distribution of the peripheral nerve. The con- in the affected nerve.
dition requires neurophysiological testing, and
sometimes imaging, to exclude other causes such
as entrapment neuropathy. Both peripheral and 4.4 Radiculoplexus Neuropathies
cranial neuropathies have a monophasic course
and resolve spontaneously; although this may Diabetic radiculoplexus neuropathies are a group
take several months. of asymmetric neuropathies with a common
Mononeuritis multiplex is a rare syndrome underlying pathophysiologic mechanism of isch-
where there is multifocal, asynchronous periph- aemic injury due to altered immunity and micro-
eral neuropathy involving at least two peripheral vasculitis [32]. They were first recognized in the
nerve trunks. There are some limited reports 19th Century and have been known by a number of
which have described the condition in patients names, including: Bruns-Garland syndrome, dia-
with diabetes [29, 30]. However, specialist input betic myelopathy, diabetic amyotrophy, femoral-­
is advised in any diabetes patient presenting with sciatic neuropathy of diabetes, etc. [33]. The
mononeuritis multiplex. condition is most well recognized to occur in dia-
betes; although a similar presentation may occur
4.3.2 Entrapment Mononeuropathies in patients without diabetes [9]. The most common
Patients with diabetes are more at risk of entrap- form is lumbosacral radiculoplexopathy, followed
ment neuropathies. These neuropathies may be by truncal radiculopathy (thoracic, abdominal or
localized to common sites of compression, e.g. thoraco-abdominal) and finally cervical radiculo-
carpal tunnel, ulnar groove, and fibula head, with plexopathy [32]. Diabetic radiculoplexus neuropa-
the median and ulnar nerve particularly prone to thies begins unilaterally in one region, but may
compression in diabetes [31]. The overweight/ spread bilaterally or cause other radiculoplexopa-
obese patient with type 2 diabetes appears to be thies, and the condition may then recur in the same
the most at risk for particularly these entrapment region. Additionally, the condition can be associ-
neuropathies. Less commonly affected nerves ated with autonomic neuropathy.
include the radial, peroneal, tibial, and lateral The condition may occur in patients with T1
cutaneous nerve of the thigh [28]. or T2DM; however, generally patients have
Entrapment neuropathies typically have a T2DM, and are in middle or old age [32, 33].
gradual onset and more persisting neuronal dys- Unlike DPN, the condition seems unrelated to
function than mononeuropathies due to micro- glycaemic control. The condition generally pres-
vasculitis. Patients may be asymptomatic or ents sub-acutely over a number of days with pain
develop paraesthesia and neuropathic pain in the in the region of the nerve distribution. The pain
Clinical Features of Diabetes Neuropathies 47

can be extreme and can have a deep or burning without the presence of a vesicular eruption.
quality. There may be associated contact allo- Neuropathy symptoms vary and may be similar
dynia. Additionally, although sensory symptoms to those of diabetic radiculoplexopathy. The
are severe, there may be a relative lack of sensory examination findings are generally of sensory
nerve dysfunction on clinical examination and disturbance, such as impairment to light touch/
neurophysiological testing. The condition is also pinprick sensation and/or allodynia/hyperalgesia,
frequently associated with weight loss, which can within the distribution of the nerve involved.
be severe, raising the concern of malignancy. Furthermore, the condition can cause local bulg-
Although pain is generally the most prominent ing of muscles within the myotome due to mus-
initial symptom, muscle weakness develops later, cular weakness.
which can be profound. The symptom course is A careful clinical history and examination can
monophasic with invariable improvement; how- easily differentiate radiculoplexus neuropathies
ever, recovery is often incomplete [33]. In par- from other diabetic neuropathies; however, confir-
ticular, muscular weakness can persist and be matory tests are necessary to confirm the diagnosis
disabling for patients and many patients require and exclude central lesions. Blood tests may reveal
lifelong walking aids, if not wheelchairs, for lum- a mild abnormality in inflammatory markers, but a
bosacral radiculoplexopathy. significant increase may suggest alternative pathol-
Lumbosacral radiculoplexopathy generally ogy. Nerve conduction studies and electromyelo-
causes symptoms in the hip and thigh, although gram abnormalities can localize the denervation.
the foot and leg may be affected. The quadriceps MRI is necessary to exclude spinal cord pathology
are classically profoundly wasted, with hip flex- (e.g., cord/nerve root compression, malignancy or
ors and abductors also commonly involved. Other demyelination) and full body cross-sectional imag-
muscles such as the adductors, glutei, and ham- ing may be considered to exclude occult malig-
strings are less commonly impacted. When there nancy. If performed, the cerebro-spinal fluid protein
is motor involvement the knee reflex is reduced level may be significantly elevated.
or absent.
Cervical radiculoplexus neuropathies are
much less common than other diabetic radiculo- 5 Conclusions
plexus neuropathies. However, a retrospective
study has systematically reviewed the features of The diabetic neuropathies are a heterogenous
condition in 85 patients, including the clinical group of disorders which can lead to great mor-
symptoms and signs [34]. The presentation may bidity, and even mortality. DPN, the commonest
be more acute than lumbosacral disease (within neuropathic disorder in diabetes, may have a sur-
24 h), with maximal deficits within 1-week. The prisingly varied clinical presentation from patient
pain was described as hurting/aching/pressure, to patient. A detailed history and clinical exami-
burning or shooting/lancinating with numbness nation with a few simple blood tests is normally
and numbness also often present. Weight loss and sufficient to diagnose this condition. However,
contralateral spread were less common than with although the clinical features of other neuropathic
cervical radiculoplexus neuropathy. Weakness syndromes affecting the cranial/peripheral nerves
was a predominant problem, with involvement in may be easily delineated with careful history and
each one of the three trunks of the brachial plexus examination, more detailed investigations, such
to a similar extent and all three involved in around as cerebral/spinal imaging and/or nerve conduc-
a third of patients. tion studies are often necessary to exclude more
Diabetic truncal radiculopathy presents with sinister pathology. Finally, although autonomic
band-like sensory symptoms within a truncal der- neuropathy is a common occurrence it generally
matome, from the back radiating anteriorly [32]. only presents clinically with advanced disease.
The symptoms are not dissimilar from the initial These conditions are difficult to diagnose, requir-
presentation of herpes zoster radiculitis; although,
48 G. Sloan et al.

ing exclusion of other causes and detailed investi- characteristics and diagnostic issues. J Diabetes
Investig. 2019;10:1148–57.
gations with specialist input. 12. Truini A, Spallone V, Morganti R, Tamburin S, Zanette
G, Schenone A, De Michelis C, Tugnoli V, Simioni V,
Manganelli F, Dubbioso R, Lauria G, Lombardi R,
References Jann S, De Toni FL, Tesfaye S, Fiorelli M, Spagnoli A,
Cruccu G.  A cross-sectional study investigating fre-
quency and features of definitely diagnosed diabetic
1. Tesfaye S, Boulton AJ, Dyck PJ, Freeman R, Horowitz
painful polyneuropathy. Pain. 2018;159:2658–66.
M, Kempler P, Lauria G, Malik RA, Spallone V,
13. Reiber GE, Vileikyte L, Boyko EJ, del Aguila M,
Vinik A, Bernardi L, Valensi P.  Diabetic neuropa-
Smith DG, Lavery LA, Boulton AJ.  Causal path-
thies: update on definitions, diagnostic criteria, esti-
ways for incident lower-extremity ulcers in patients
mation of severity, and treatments. Diabetes Care.
with diabetes from two settings. Diabetes Care.
2010;33:2285–93.
1999;22:157–62.
2. Ziegler D, Papanas N, Vinik AI, Shaw
14. Shillo P, Sloan G, Greig M, Hunt L, Selvarajah D,
JE. Epidemiology of polyneuropathy in diabetes and
Elliott J, Gandhi R, Wilkinson ID, Tesfaye S. Painful
prediabetes. Handb Clin Neurol. 2014;126:3–22.
and painless diabetic neuropathies: what is the differ-
3. Andersen ST, Witte DR, Dalsgaard EM, Andersen
ence? Curr Diab Rep. 2019;19:32.
H, Nawroth P, Fleming T, Jensen TM, Finnerup NB,
15. Themistocleous AC, Ramirez JD, Shillo PR, Lees
Jensen TS, Lauritzen T, Feldman EL, Callaghan
JG, Selvarajah D, Orengo C, Tesfaye S, Rice AS,
BC, Charles M.  Risk factors for incident diabetic
Bennett DL. The pain in neuropathy study (PiNS): a
polyneuropathy in a cohort with screen-detected
­
cross-sectional observational study determining the
type 2 diabetes followed for 13 years: ADDITION-
somatosensory phenotype of painful and painless dia-
Denmark. Diabetes Care. 2018;41:1068–75.
betic neuropathy. Pain. 2016;157:1132–45.
4. Tesfaye S, Chaturvedi N, Eaton SE, Ward JD,
16. Ward JD.  The diabetic leg. Diabetologia.
Manes C, Ionescu-Tirgoviste C, Witte DR, Fuller
1982;22:141–7.
JH. Vascular risk factors and diabetic neuropathy. N
17. Galer BS, Gianas A, Jensen MP. Painful diabetic poly-
Engl J Med. 2005;352:341–50.
neuropathy: epidemiology, pain description, and qual-
5. Pop-Busui R, Boulton AJ, Feldman EL, Bril
ity of life. Diabetes Res Clin Pract. 2000;47:123–8.
V, Freeman R, Malik RA, Sosenko JM, Ziegler
18. Gylfadottir SS, Christensen DH, Nicolaisen SK,
D.  Diabetic neuropathy: a position statement by
Andersen H, Callaghan BC, Itani M, Khan KS,
the American Diabetes Association. Diabetes Care.
Kristensen AG, Nielsen JS, Sindrup SH, Andersen
2017;40:136–54.
NT, Jensen TS, Thomsen RW, Finnerup NB. Diabetic
6. Rota E, Zavaroni D, Parietti L, Iafelice I, De Mitri P,
polyneuropathy and pain, prevalence, and patient
Terlizzi E, Morelli N, Immovilli P, Guidetti D. Ulnar
characteristics: a cross-sectional questionnaire study
entrapment neuropathy in patients with type 2 diabe-
of 5,514 patients with recently diagnosed type 2 dia-
tes mellitus: an electrodiagnostic study. Diabetes Res
betes. Pain. 2020;161:574–83.
Clin Pract. 2014;104:73–8.
19. Sadosky A, Mardekian J, Parsons B, Hopps M,
7. Dyck PJ, Kratz KM, Karnes JL, Litchy WJ, Klein R,
Bienen EJ, Markman J.  Healthcare utilization and
Pach JM, Wilson DM, O’Brien PC, Melton LJ.  The
costs in diabetes relative to the clinical spectrum of
prevalence by staged severity of various types of dia-
painful diabetic peripheral neuropathy. J Diabetes
betic neuropathy, retinopathy, and nephropathy in
Complications. 2015;29:212–7.
a population-based cohort: the Rochester Diabetic
20. Gore M, Brandenburg NA, Dukes E, Hoffman DL, Tai
Neuropathy Study. Neurology. 1993;43:817–24.
KS, Stacey B. Pain severity in diabetic peripheral neu-
8. Greco D, Gambina F, Pisciotta M, Abrignani M,
ropathy is associated with patient functioning, symp-
Maggio F.  Clinical characteristics and associated
tom levels of anxiety and depression, and sleep. J Pain
comorbidities in diabetic patients with cranial nerve
Symptom Manage. 2005;30:374–85.
palsies. J Endocrinol Invest. 2012;35:146–9.
21. Papanas N, Ziegler D. Risk factors and comorbidities
9. Ng PS, Dyck PJ, Laughlin RS, Thapa P, Pinto MV,
in diabetic neuropathy: an update 2015. Rev Diabet
Dyck PJB.  Lumbosacral radiculoplexus neuropathy:
Stud. 2015;12:48–62.
incidence and the association with diabetes mellitus.
22. Dietrich I, Braga GA, de Melo FG, da Costa Silva
Neurology. 2019;92:1188–94.
AC.  The diabetic foot as a proxy for cardiovascular
10. Rajabally YA, Stettner M, Kieseier BC, Hartung HP,
events and mortality review. Curr Atheroscler Rep.
Malik RA.  CIDP and other inflammatory neuropa-
2017;19:44.
thies in diabetes  - diagnosis and management. Nat
23. Gibbons CH, Freeman R. Treatment-induced neurop-
Rev Neurol. 2017;13:599–611.
athy of diabetes: an acute, iatrogenic complication of
11. Gylfadottir SS, Weeracharoenkul D, Andersen ST,
diabetes. Brain. 2015;138:43–52.
Niruthisard S, Suwanwalaikorn S, Jensen TS. Painful
24. Gibbons CH. Treatment induced neuropathy of diabe-
and non-painful diabetic polyneuropathy: Clinical
tes. Auton Neurosci. 2020;226:102668.
Clinical Features of Diabetes Neuropathies 49

25. Gibbons CH.  Treatment induced neuropathy of 30. Kelkar P, Parry GJ. Mononeuritis multiplex in diabetes
diabetes-­long term implications in type 1 diabetes. J mellitus: evidence for underlying immune pathogen-
Diabetes Complications. 2017;31:715–20. esis. J Neurol Neurosurg Psychiatry. 2003;74:803–6.
26. Spallone V. Update on the impact, diagnosis and man- 31. Rota E, Morelli N. Entrapment neuropathies in diabe-
agement of cardiovascular autonomic neuropathy in tes mellitus. World J Diabetes. 2016;7:342–53.
diabetes: what is defined, what is new, and what is 32. Laughlin RS, Dyck PJ. Diabetic radiculoplexus neu-
unmet. Diabetes Metab J. 2019;43:3–30. ropathies. Handb Clin Neurol. 2014;126:45–52.
27. Vinik AI, Maser RE, Mitchell BD, Freeman 33. Dyck PJ, Windebank AJ.  Diabetic and nondiabetic
R.  Diabetic autonomic neuropathy. Diabetes Care. lumbosacral radiculoplexus neuropathies: new
2003;26:1553–79. insights into pathophysiology and treatment. Muscle
28. Vinik A, Mehrabyan A, Colen L, Boulton A.  Focal Nerve. 2002;25:477–91.
entrapment neuropathies in diabetes. Diabetes Care. 34. Massie R, Mauermann ML, Staff NP, Amrami KK,
2004;27:1783–8. Mandrekar JN, Dyck PJ, Klein CJ.  Diabetic cervi-
29. Raff MC, Sangalang V, Asbury AK. Ischemic mono- cal radiculoplexus neuropathy: a distinct syndrome
neuropathy multiplex associated with diabetes melli- expanding the spectrum of diabetic radiculoplexus
tus. Arch Neurol. 1968;18:487–99. neuropathies. Brain. 2012;135:3074–88.
Neuropathy in Type 1 and Type 2
Diabetes

Gulcin Akinci, Dustin Nowacek,
and Brian Callaghan

1 Introduction have other metabolic syndrome components,


whereas type 1 diabetes does not cluster as fre-
Diabetic peripheral neuropathy (DPN) is charac- quently with these comorbidities. Furthermore,
terized by injury to peripheral somatic and/or type 1 diabetes often starts very early in life,
autonomic nerves [1]. DPN is also one of the whereas the prevalence of type 2 diabetes
most common chronic complications of diabetes increases substantially as age increases. Given
affecting up to 50% of people living with diabe- these major difference between type 1 and type 2
tes (Table 1) [2, 3]. In addition to a high preva- diabetes, it is not surprising that many differences
lence, DPN causes substantial morbidity. are also seen in the neuropathy that results from
Specifically, patients with DPN often suffer from these two conditions.
severe pain, reduced quality of life, and frequent Among DPN, distal symmetrical polyneurop-
falls [4, 5]. Diabetic foot ulcers and amputation athy (DSP) is the most common pattern of nerve
are also devastating complications of this com- injury, thus DSP will be the main focus of this
mon condition. While type 1 diabetes and type 2 chapter, but we will also discuss other patterns of
diabetes can both lead to DPN, these two condi- nerve injury (Fig.  1) [6]. Furthermore, we will
tions differ in substantial ways. Type 1 diabetes is cover clinical features, risk factors, and clinical
caused by a lack of insulin, whereas type 2 diabe- trials in diabetic neuropathy. We will also high-
tes is caused by several metabolic abnormalities. light the differences between neuropathy in those
In addition, patients with type 2 diabetes often with type 1 and type 2 diabetes.

G. Akinci
Department of Neurology, University of Michigan,
Ann Arbor, MI, USA
Department of Pediatrics, Division of Pediatric
Neurology, Koc University, Istanbul, Turkey
D. Nowacek · B. Callaghan (*)
Department of Neurology, University of Michigan,
Ann Arbor, MI, USA
e-mail: bcallagh@med.umich.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 51


S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_4
52 G. Akinci et al.

Table 1  The incidence of polyneuropathies, mononeuropathies, and radiculopathies


Polyneuropathy Population studied Incidence per 100,000 person years
All causes Netherlands [7] 77.0
Type 1 diabetes USA [8] 2800
Type 2 diabetes USA [9] 6100
Mononeuropathies Population studied Incidence per 100,000 person years
Median neuropathy at the Wrist UK [10, 11] 103 (men 87.8, women 192.8)
(carpal tunnel syndrome) USA [12] 99
Ulnar neuropathy UK [11] Men 25.2, women 18.9
Italy (Siena) [13] 24.7 (men 32.7, women 17.2)
Lateral femoral cutaneous UK [11] Men 10.7, women 13.2
neuropathy (meralgia paresthetica) Netherlands [14] 43
Radial neuropathy UK [11] Men 2.97, women 1.42
Idiopathic facial neuropathy (bell’s UK [15] 20.2
palsy) USA (Rochester, MN) [16] 25 (men 22.8, women 26.9)
Italy (Rome) [17] 53.3
Radiculopathies Population studied Incidence per 100,000 person years
Lumbar USA Military [18] 486 (1,079 in patients over 40)
Cervical USA Military [19] 179 (616 in patients over 40)
USA (Rochester, MN) [20] 83.2 (202.9 in patients 50–54; men 107.3,
women 63.5)
USA United States of America, UK United Kingdom

a c

b d

Fig. 1  Patterns of nerve injury in diabetic neuropathy. (a) distal symmetric polyneuropathy; (b) diabetic radiculopathy
and radiculoplexus neuropathy; (c) multiple mononeuropathies; (d) autonomic neuropathy
Neuropathy in Type 1 and Type 2 Diabetes 53

2 Clinical Features of DPN tective sensation, and progressive numbness.


Other symptoms of late-stage DSP include distal
DPN can involve multiple patterns of nerve motor nerve involvement resulting in muscle
injury; therefore, symptoms can vary signifi- weakness, unsteadiness, imbalance and gait
cantly. These wide ranging neuropathic symp- problems, and impaired proprioception [29].
toms have been recognized in patients with DSP has a similar prevalence in type 1 and
diabetes for more than a hundred years and the type 2 diabetes [30]. Despite similar clinical fea-
earliest classifications of DPN divided patients tures in DSP in those with type 1 and type 2 dia-
into three groups: hyperesthetic, paralytic, and betes, the natural history among subjects with
ataxic forms of neuropathy [21]. The most recent these two major types of diabetes differs signifi-
Position Statement of American Diabetes cantly highlighting the fundamental differences
Association classifies neuropathies based on in disease pathophysiology [31]. Type 1 diabetes
which parts of peripheral nervous system are is caused by absolute insulin deficiency; how-
affected and includes diffuse neuropathies (DSP ever, the pathophysiology of type 2 diabetes is
and autonomic neuropathies), mononeuropa- multifactorial and quite complex involving a con-
thies, and radiculopathies [6]. Most studies have stellation of metabolic insults including insulin
focused on DSP and cardiovascular autonomic resistance [32]. Many clinical presentations of
neuropathy (CAN) because they are the most diabetic neuropathy overlap in patients with type
common manifestations. 1 and type 2 diabetes. However, a few observa-
DSP, a chronic, symmetrical, length-­tional studies give us insight into clinical differ-
dependent sensorimotor polyneuropathy, is the ences in neuropathy presentations between these
most common pattern of DPN [3]. DSP is a sen- major types of diabetes. Symptomatic DSP typi-
sorimotor neuropathy in which sensory ­symptoms cally develops in patients with type 1 diabetes
are predominant at the early stages of the disease after a long disease duration and poorly con-
[6]. DSP is a mixed polyneuropathy that usually trolled hyperglycemia. On the other hand, this
starts with small fiber neuropathy and progresses presentation may be less common in patients
to large fiber neuropathy [22, 23]. Small fibers with type 2 diabetes. Patients with type 2 diabetes
involve predominantly small diameter somatic can develop neuropathy even within a few years
and autonomic nerves, unmyelinated C fibers, of their diabetes diagnosis and some patients may
and/or thinly myelinated A-delta sensory fibers. even exhibit signs and symptoms at the time of
They carry information regarding pain and tem- diabetes diagnosis [33]. Even prior to diabetes
perature and also innervate the autonomic ner- onset, patients with prediabetes may present with
vous system. Large fibers are myelinated fibers painful DSP as the first symptom of carbohydrate
that relay vibratory and proprioceptive informa- intolerance [34, 35].
tion. Symptoms are varied by the class of nerve Multiple studies have investigated differences
fiber affected. The most common early symptoms in the natural history of DSP between those with
related to small fiber involvement include pain type 1 and type 2 diabetes. The Oslo study
and dysesthesia [24]. About one-­third of patients showed that small fiber neuropathy dominates the
with DPN may experience neuropathic pain [25] clinical findings of patients with longstanding
that worsens at rest and may show some relief type 1 diabetes and that the nerve injury is cor-
while walking [26]. Pain can be spontaneous or related with glycemic control [36]. In another
triggered by external stimuli (allodynia and study, subjects with latent autoimmune diabetes
hyperalgesia). Painful neuropathy can be dis- of adulthood (LADA), who had worse glycemic
abling and may persist for years [27]. On some control, were found to have greater small fiber
occasions, pain may diminish after sensory loss involvement than subjects with type 2 diabetes
worsens [28]. As DSP progresses, large nerve despite showing a more favorable BMI and lipid
fibers are affected, resulting in additional symp- profile [37]. Although the frequency and clinical
toms such as tingling without pain, loss of pro- presentation of neuropathy were similar in sub-
54 G. Akinci et al.

jects with type 1 and type 2 diabetes (described in subjects with type 2 diabetes [44–46].
as IDDM and NIDDM at that time) in the Nonspecific symptoms such as fatigue, general
Rochester Diabetic Neuropathy Study [38], neu- weakness, and dizziness may be present. The
ropathy was significantly more severe in type 1 prevalence of CAN reaches up to 50% after 15
diabetes as staged by the investigators systemati- years of type 2 diabetes [6]. Although CAN is
cally. However, it should be noted that patients unusual in newly diagnosed patients with type 1
with type 1 diabetes had a longer duration of dia- diabetes, studies have shown that it develops in
betes, which may account for at least some of this up to 30% of them after 20 years of diagnosis of
difference. Loseth et al. [39], on the other hand, diabetes [47, 48].
reported that small fiber neuropathy progressed Gastrointestinal presentations of DAN appear
more rapidly in patients with type 2 diabetes to be more common in patients with type 1 diabe-
compared to those with type 1 diabetes after 5 tes but can also be observed in patients with long-
years of follow-up, even though both groups had standing type 2 diabetes [49, 50]. These
similar neuropathy prevalence in an earlier cross-­ presentations are typically observed in patients
sectional study of the same cohort [40]. Also, a with poorly controlled glycemia and in the pres-
large observational study from England detected ence of other microvascular complications of dia-
painful neuropathic symptoms more commonly betes. Diabetic gastroparesis causes delayed
in subjects with type 2 diabetes than type 1 diabe- gastric emptying that can trigger brittle diabetes
tes, highlighting the presence of painful neuropa- that in turn may eventually result in poorer glyce-
thy symptoms in relatively early years of diabetes mic control. In addition, gastroparesis often leads
in type 2 diabetes [25]. Although differences in to symptoms such as nausea, vomiting, early sati-
fiber type involvement, severity, and pain have ety, and abdominal pain. Gastrointestinal dys-
been demonstrated between type 1 and type 2 motility is not limited to the upper gastrointestinal
diabetes, the similarities far outweigh the system in diabetes and can affect small bowels
differences. causing constipation and episodes of diarrhea
The definition of diabetic autonomic neuropa- [51]. Similar to gastroparesis, studies suggest
thy (DAN) covers several different clinical pre- that diabetic diarrhea may be more common in
sentations ranging from cardiovascular autonomic patients with type 1 diabetes [52, 53]. Several
neuropathy (CAN) to diabetic gastroparesis, conditions should be considered in diabetic
hypoglycemia unawareness, and sweating dys- patients presenting with episodes of diarrhea that
regulation [6, 41]. Besides a variety of clinical include, but are not limited to, bacterial over-
presentations, the severity of DAN is also hetero- growth, exocrine pancreatic deficiency, coexist-
geneous. Among different presentations of DAN, ing Celiac disease, and metformin use [52, 53].
CAN is one of the most serious complication and DAN can also affect genitourinary system.
relatively more comprehensively studied in the Clinical presentations may vary from bladder
literature. CAN has been shown to be an indepen- dysfunction to sexual dysfunction [54]. Erectile
dent risk factor for cardiovascular mortality in dysfunction (ED) is a relatively common prob-
type 1 and type 2 diabetes [6, 42, 43]. Although lem that can develop in males with both type 1
the exact prevalence of CAN is still unknown, a and type 2 diabetes and is associated with a
previous study showed that roughly one-fourth of higher incidence of cardiovascular disease [55,
patients with type 1 and one-third of subjects 56]. Incidence of ED is higher in diabetic males
with type 2 had signs of CAN when screened with metabolic syndrome [57]. Diabetic bladder
[44]. Early stages of CAN are usually asymptom- dysfunction, on the other hand, more commonly
atic but can be still detected by simple screening develops in patients with type 1 diabetes [54].
tests such as heart rate variability (HRV) tests. Patients with diabetic bladder may not sense a
Studies have shown that HRV tests can detect full bladder due to afferent nerve involvement or
abnormalities within 2 years in patients with type may suffer from incomplete emptying as a result
1 diabetes and earlier (even at prediabetic stage) of efferent nerve involvement. Clinical conse-
Neuropathy in Type 1 and Type 2 Diabetes 55

quences may include urinary incontinence, poor Diabetic amyotrophy, a disabling proximal neu-
urinary stream, dribbling, and recurrent urinary ropathy affecting lower extremities, can develop
tract infections. Urinary incontinence due to in both type 1 and type 2 diabetes with a rela-
DAN is classically associated with type 1 diabe- tively higher prevalence in type 2 diabetes [62].
tes and is relatively less common among patients The pathophysiology of diabetic amyotrophy is
with type 2 diabetes [58, 54]. Analogous to DSP, not well understood; however, relatively slower
differences in DAN between patients with type 1 progression after rapid onset of disease suggests
and type 2 diabetes exist, but the similarities out- both metabolic and vascular involvement [63].
weigh these differences. Likewise, mononeuropathies and other radicu-
Acute painful neuropathy of poor glycemic lopathies are more commonly reported in type 2
control is a relatively rare clinical presentation diabetes. Although rare, cranial mononeuropa-
that develops in subjects with poorly controlled thies (especially oculomotor nerve palsy) can be
diabetes and is usually associated with severe observed in patients with diabetes. These patients
weight loss [59]. It is not clear if weight loss is are typically relatively older individuals with
due to insulin deficiency but neuropathic pain longstanding poorly controlled type 2 diabetes
improves after weight gain following insulin with other complications of diabetes; however,
treatment. Typical patients are poorly controlled oculomotor nerve palsy has been reported in type
males with type 2 diabetes who are on oral anti- 1 diabetes as well [64]. Typically, diabetes affects
diabetics (or newly diagnosed) but it can develop the central portion of the oculomotor nerve with-
in patients with type 1 diabetes as well. Pain is out other cranial nerve involvements, causing
usually severe and often leads to depressive deviation of the eye with normal pupil reactions
symptoms. Another form of acute painful neu- [65]. However, other causes of oculomotor nerve
ropathy can occur after rapid glycemic control. palsy such as intracranial tumors, aneurysms, and
This phenomenon had probably been underre- brainstem strokes should be considered in differ-
ported as recent observations suggest a higher ential diagnosis [66]. Finally, entrapment neu-
prevalence than previously thought. Recently, the ropathies are quite common in people with
term “treatment-induced neuropathy of diabetes” diabetes, median neuropathy being the most
(TIND) has been suggested to describe this con- common presentation [67].
dition [60]. Rapid correction of glycemic control
can trigger acute/subacute painful neuropathy
that more commonly happens in poorly con- 3 Risk Factors for DPN
trolled patients with type 1 diabetes but it can
also be observed in patients with type 2 diabetes The pathogenesis of DPN is complex with a wide
[61]. Several interesting recent observations range of risk factors being involved (Fig.  2).
regarding TIND include higher prevalence of eat- Independent of the patient’s age, diabetes dura-
ing disorders among individuals with type 1 dia- tion is a risk factor in both types of diabetes type
betes that developed TIND and the occurrence of [68–71]. In addition to traditional risk factors
symptoms mostly in older subjects with type 2 such as age and disease duration, hyperglycemia
diabetes [60]. The key is to avoid rapid drops in is the most widely studied risk factor; however,
hyperglycemia and target more gradual glycemic current evidence suggests novel metabolic risk
control in patients experiencing TIND. factors are important in the pathogenesis of DPN
Patients with diabetes may present with focal [72].
and multifocal neuropathies such as diabetic Current evidence supports a closer relation-
amyotrophy, cranial neuropathy, and limb and ship between glucose control and DPN in type 1
truncal neuropathies, although these clinical pre- diabetes than type 2 diabetes [73]. Hyperglycemia
sentations are far less common than DSPs. These is a robust risk factor for DPN in type 1 diabetes,
neuropathies are typically observed predomi- and current literature supports the role of good
nantly in older patients with type 2 diabetes. glycemic control that would reduce the incidence
56 G. Akinci et al.

Fig. 2  Signaling pathways underlying nutrient excess and metabolic neuropathy

of DPN [47, 74, 75]. Also, the occurrence and On the other hand, there is a weaker correla-
progression of DPN are more predictable in type tion between DPN and glycemic status in type 2
1 diabetes. DPN typically becomes symptomatic diabetes. Type 2 diabetes is usually complicated
after years of chronic poor glycemic control and with multiple comorbidities that might affect
progresses unless glycemic control is achieved DPN outcomes and have attenuated the benefit of
[76]. glycemic control. Most patients with type 2 dia-
Previous studies found a significant benefit of betes are overweight or obese, have dyslipidemia,
intensive glycemic control to improve DPN hypertension, and a high cardiometabolic risk
related outcomes in patients with type 1 diabetes. profile. In addition, contrary to observations in
The landmark Diabetes Control and Complications type 1 diabetes, DPN can be relatively common
Trial (DCCT) study reported that the risk of DPN among subjects with newly diagnosed diabetes
and CAN decreased by 64% and 45% after inten- [68]. Patients may be diagnosed with DPN at the
sive glycemic control [66]. This benefit remained same time when they are diagnosed with diabetes
significant in the observational follow-up of this [81]. Signs of DPN can be detected in prediabetic
cohort [47, 77]. Glycemic improvement was also conditions (in the absence of overt diabetes) such
associated with lower DPN incidence in the as impaired fasting glucose/ impaired glucose
Pittsburgh Epidemiology of Diabetes tolerance [82–84], obesity [85–90], and meta-
Complications (EDC) study [78]. Similar trends bolic syndrome [85, 91].
were observed in the European Diabetes The benefit of glycemic control in type 2 dia-
Prospective Complications (EURODIAB) study betes is less conclusive. Although benefits of bet-
[79] cohort, and the recent SEARCH study con- ter glycemic control have been reported in several
firmed this association in youth with type 1 diabe- studies in type 2 diabetes [92, 93], several others
tes [80]. In parallel to these evidence, our previous have failed to show the benefit of glycemic con-
meta-analysis showed that optimized glycemic trol on DPN outcomes [94–97]. Our previous
control in type 1 diabetes reduces the develop- meta-analysis failed to show a significant benefit
ment of clinical DPN [73]. of glycemic control on DPN, although the short-
Neuropathy in Type 1 and Type 2 Diabetes 57

age of primary outcome studies in type 2 diabetes common among subjects with type 2 diabetes
constitutes a limitation [73]. Several studies have than those with type 1 diabetes. Recent studies
also reported an association between glycemic found a significant relationship between compo-
variability and neuropathy in type 1 and type 2 nents of metabolic syndrome and DPN [85, 86,
diabetes [98–100]; however, this association was 88, 89, 104, 110, 111]. Besides obesity, emerging
not observed in the DCCT analyses [98, 101]. data suggest a possible association between other
Also, further small-scale studies failed to confirm components of metabolic syndrome and DPN as
this association [102]. well, although this association seems to be less
Obesity is a major risk factor for type 2 diabe- consistent [72]. In the general population, the
tes that plays a fundamental role in the disease likelihood of DPN is increased in subjects with
pathogenesis. Obesity contributes to a higher risk metabolic syndrome [112]. DPN prevalence was
of diabetes complications. The association also reported to increase in parallel to the increas-
between obesity and typical conditions that are ing number of metabolic syndrome components
commonly associated with diabetes such as car- [85, 113]. Insulin resistance, the core shared
diovascular diseases or nephropathy is robust, pathophysiology in type 2 diabetes and metabolic
even in the absence of diabetes [103]. Multiple syndrome, has been identified as a major inde-
studies have demonstrated that obesity is consis- pendent risk factor of DPN [114].
tently associated with neuropathy [85, 88–90, Dyslipidemia appears to be a contributing
104]. Based on these studies, obesity has been factor to DPN [72]. Many patients with type 2
proposed as an independent risk factor for dia- diabetes exhibit elevated triglycerides to some
betic neuropathy [105]. Our previous studies degree. A recent meta-analysis reported a higher
have shown a higher prevalence of DPN in obese risk of DPN among people with increasing lev-
subjects with normoglycemia than in lean con- els of triglycerides [115]. The Utah Diabetic
trols in two different cohorts [86, 88]. We also Neuropathy Study reported that hypertriglyceri-
found that CAN, along with DPN, was a common demia increased the risk of DPN in patients with
complication in patients with severe obesity [86]. type 2 diabetes and triglyceride levels were
Higher BMI was associated with reduced HRV associated with loss of small unmyelinated
and parasympathetic loss and sympathetic over- axons [113]. Hypertriglyceridemia was identi-
drive in adolescents and youth with type 2 diabe- fied as an independent risk factor for lower-
tes in the TODAY [106]. Our work and others extremity amputations in diabetic patients in the
also revealed the role of central obesity in DPN 10-year follow-up of the DISTANCE study
[86, 107]. Therefore, obesity has emerged as the cohort [116]. Adolescents and young adults
second most important metabolic risk factor for with type 2 diabetes and CAN had significantly
neuropathy. higher triglyceride levels in the SEARCH study
Although obesity is usually attributed to type [45]. A similar association was also reported in
2 diabetes, it is being increasingly reported in patients with type 2 diabetes from the Anglo-
type 1 diabetes populations as well [108]. Similar Danish-Dutch study of Intensive Treatment of
to type 2 diabetes, several reports indicate asso- Diabetes in Primary Care (ADDITION)-
ciations between metabolic abnormalities that Denmark study [117]. The ADDITION study
link to obesity and DPN among subjects with reported lower HDL cholesterol levels as a risk
type 1 diabetes. Indexes of obesity have been factor for diabetic neuropathy [104]. An inde-
reported to be associated with diabetic neuropa- pendent significant correlation between low
thy in type 1 diabetes in several studies [79, 80, HDL cholesterol and higher diabetic DPN risk
109]. was also reported in T2D youths within the
Metabolic syndrome is a cluster of conditions SEARCH cohort [80].
that has been closely associated with insulin Dyslipidemia has previously been proposed as
resistance and the risk of type 2 diabetes. The a risk factor for diabetic neuropathy in subjects
prevalence of metabolic syndrome is much more with type 1 diabetes as well [69, 75, 80]. In paral-
58 G. Akinci et al.

lel to these studies, elevated triglycerides were 4 Clinical Trials in DPN


found to be a significant risk factor for DPN in
the recently published report of long-term fol- Several clinical trials have assessed the effect of
low-­up (>23 years) of the DCCT-EDIC cohort glucose control on the development of polyneu-
[71]. The study also found a strong independent ropathy in patients with diabetes mellitus. A 2012
association between hypertension and DPN, sup- Cochrane review identified these studies and ana-
porting previously published results from the lyzed them in a meta-analysis (Table  2). In this
Pittsburgh EDC [75], EURODIAB [79], and review, the primary outcome was incidence of
SEARCH studies [80]. clinical neuropathy in patients without clinical
Finally, hypertension, a major components neuropathy at baseline. Secondary outcomes
of metabolic syndrome, has been previously comprised of changes in neuropathic symptoms,
found to be associated with diabetic neuropathy nerve conduction studies (NCS) and quantitative
[45]. Although a link between hypertension and sensory testing, and adverse events. Adverse
DPN has been reported in patients with type 2 events consisted of foot ulcers, amputations, and
diabetes [118, 119], the association of hyper- hypoglycemic episodes requiring hospitalization.
tension with DPN has been more clearly shown Outcomes measured after less than 12 months
among subjects with type 1 diabetes in multiple were not included. Seven clinical trials evaluated
studies [71, 75, 79, 8, 120]. While hyperglyce- people with type 1 diabetes, eight clinical trials
mia remains the most important metabolic risk included people with type 2 diabetes mellitus, and
factor for DPN, all of the other metabolic syn- two trials included patients with both type 1 and
drome components are associated with type 2 diabetes. In the trials that studied exclu-
DPN.  While the patients with type 2 diabetes sively type 1 diabetes, most compared different
are more likely to have these comorbidities, insulin regimens, and the two studies that included
they are also associated with DPN in those with both types of diabetes also compared different
type 1 diabetes. insulin regimens. Of the eight studies that evalu-
ated type 2 diabetes, three evaluated different

Table 2  Clinical trials investigating the role of enhanced glucose control on neuropathy
Trial Length of study Clinical Other Enhanced glycemic control
Investigator size (years) outcome outcomes superior?
Type 1 diabetes
Holman (1983) 74 2 No QST Yes
Lauritzen (1985) 30 2 No QST No
Dahl-Jorgensen 45 2 No NCS Yes
(1986)
Jakobsen (1988) 24 2 No QST Yes
DCCT (1993) 1441 5 Yes NCS Yes
Reichard (1993) 102 7.5 No NCS, QST Yes
Linn (1996) 49 5 Yes None Yes
Type 2 diabetes
Kawamori (1991) 50 4 No NCS Yes
Ohkubo (1995) 110 6 No NCS, QST Yes
UKPDS (1998) 3867 10 No QST Yes
Tovi (1998) 38 1 Yes None No
Azad (1999) 153 2 Yes None No
Shichiri (2000) 110 8 No NCS, QST Yes
Gaede (2003) 160 8 No QST No
Duckworth (2009) 1791 5.6 Yes None No
ACCORD (2010) 10,251 3.7 Yes None No
Neuropathy in Type 1 and Type 2 Diabetes 59

insulin regimens and five evaluated other treat- reduction after 5 years. In the intensive treatment
ment strategies, including more aggressive glyce- groups in both groups combined there was a risk
mic goals through the use of diet and exercise, reduction of 1.74% per year (95% CI 1.00–2.48)
oral hypoglycemic agents, insulin, or oral hypo- [73, 118, 119].
glycemic agents plus insulin. The primary out- The other study reporting the primary out-
come was reported in two of the studies involving come of incident neuropathy was Linn et  al.,
type 1 diabetes [118–121], and in four of the stud- which randomized 49 consecutive participants
ies involving type 2 diabetes [93, 95, 122, 123]. with newly diagnosed insulin dependent diabetes
The largest study in type 1 diabetes was the mellitus (IDDM) to intensive or conventional
DCCT [118, 119], which randomized 1441 par- insulin therapy, and evaluated them 5 years after
ticipants. Primary prevention participants (N = their clinical diagnosis. Definite neuropathy was
726) were defined as having insulin dependent defined as the presence of three of the following:
diabetes mellitus of 1–5 years’ duration, no symptoms, examination signs, abnormal quanti-
detectable retinopathy on stereo fundus photog- tative sensory testing, and peroneal motor nerve
raphy, and urinary albumin excretion less than 40 conduction velocity. Intensive therapy included
mg/24 h. Secondary prevention participants (N = administration of insulin at least three times daily
715) were defined as having insulin dependent by injection with dosage being adjusted based on
diabetes mellitus of 1–15 years’ duration, very monitoring of blood glucose, dietary intake, and
mild to moderate non-proliferative retinopathy, anticipated exercise. Target blood glucose in the
and urinary albumin excretion less than 200 intensive group was defined as self-determined
mg/24 h. Almost all participants (N = 1436) had a capillary glucose less than 6.8 mmol/L and less
baseline neuropathy assessment). Those in the than 10 mmol/L postprandially, and a hemoglo-
intensive group received either an external insu- bin A1c less than 6.5. Conventional therapy con-
lin pump, or 3 or more daily insulin injections sisted of 1 or 2 daily injections of insulin
guided by frequent blood glucose monitoring. including mixed intermediate and rapid-acting
Participants in the conventional group received 1 insulin and variable self-monitoring of blood glu-
or 2 daily insulin injections. The goals of the cose. The goals of conventional therapy included
intensive group included a pre-prandial blood the absence of symptoms attributed to glucosuria
glucose between 70 and 120 mg/dL, postprandial or hyperglycemia, and freedom from severe or
levels less than 180 mg/dL, a weekly 3 AM mea- frequent hypoglycemia. After 5 years, 1 partici-
surement greater than 65 mg/dL, and a monthly pant in the intensive group and 6 participants in
HgbA1c less than 6.05%. Definite neuropathy the conventional group developed neuropathy.
was defined as the presence of two or more of the This study was much smaller than the DCCT
following: symptoms, sensory examination find- study, but notably it revealed a 5.45% per year
ings, and decreased or absent reflexes, and hav- risk reduction (95% CI 0.95–9.95) and an 86%
ing one of these abnormalities was defined as relative risk reduction in clinical neuropathy
“possible” neuropathy, and “confirmed” definite [121]. The meta-analysis including the DCCT
clinical neuropathy also required the finding of and Linn studies revealed a statistically signifi-
an unequivocal abnormality on nerve conduction cant 1.84% per year risk reduction (95% CI 1.11–
studies or autonomic nervous system testing. 2.56) [73]. Therefore, current evidence supports
NCS were performed at baseline, at 5 years and a large effect of glycemic control on reducing
at study end. Participants were followed for a neuropathy in patients with type 1 diabetes.
mean of 6.5 years. In the primary prevention While the above two studies were the only
cohort there was 1.53% per year risk reduction studies that reported clinical neuropathy as an
(95% CI 0.51–2.54) and a relative risk reduction outcome in type 1 diabetes in the Cochrane
after 5 years of 53%. In the secondary prevention review, a majority of the five other studies that
cohort there was a 1.97% per year risk reduction evaluated the secondary outcomes described
(95% CI 0.90–3.04) and a 52% relative risk above were also in favor of the intensive treat-
60 G. Akinci et al.

ment groups. Multiple studies demonstrate an on a yearly physical exam, with no other details
annual mean difference (MD) in conduction being provided regarding what constitutes a diag-
velocity of between 0.4 and 0.6 m/s of different nosis of neuropathy. 178 of the 464 participants
motor nerves in favor of the intensive group, pro- developed neuropathy in the intensive group
viding further evidence of improvement in nerve compared with 199 of 498 participants in the con-
function in this group. Four studies in type 1 dia- ventional droop. Ultimately this resulted in a
betes evaluating quantitative vibration testing 0.29% per year risk reduction (95% CI –0.82 to
showed improvement, with 3 of the studies 1.39) and a 4% relative risk reduction at a median
reporting enough information to allow for meta-­ follow-up of 5.6 years, but these results did not
analysis, which showed an annual standardized meet statistical significance [95].
mean difference (SMD) of 0.32 (95% CI 0.02– Two other studies evaluated the primary out-
0.43) in favor of intensely treated participants. come of incident neuropathy in patients with type
Notably, there was a threefold increase in epi- 2 diabetes. Azad 1999 investigated 153 partici-
sodes of serious hypoglycemia in those receiving pants over 2 years randomized to conventional
intensive treatment. Despite this important versus enhanced treatment with insulin, which
adverse event, the substantial benefit in not only lowered the hemoglobin A1c by 2.1% lower than
neuropathy outcomes but also in other health-­ the standard arm, but there was no significant dif-
related outcomes (nephropathy and retinopathy) ference in the incidence of neuropathy between
in patients receiving intensive glycemic control the treatment groups [122]. Tovi followed 40
strongly suggests that the risk/benefit ratio is in elderly type 2 diabetic participants who had sec-
favor of intensive treatment [73]. ondary failure of oral diabetic drug therapy with-
In the 2012 Cochrane review, four studies out symptoms of hyperglycemia, and these
investigated the primary outcome in type 2 diabe- participants were randomized to insulin versus
tes. The largest study was the Accord 2010, oral hypoglycemic agents and followed for 1 year.
which randomized 10,251 participants in North Clinical neuropathy was defined as a composite
America. This study included patients with score utilizing examination and electrodiagnostic
HgbA1c greater than 7.5% and cardiovascular findings, and ultimately no patient's in either group
disease or 2 or more cardiovascular risk factors. developed neuropathy [123]. Of these two studies,
Participants were randomized to an intensive tar- there was no statistical significant difference in
get (HgbA1c less than 6.0%) or a standard target favor of either group [73]. Overall the meta-analy-
(7.0–7.9%) and followed for a median 3.7 years. sis of these four studies (Accord, Duckworth,
Clinical neuropathy was defined as a score of Azad, Tovi) revealed a risk reduction of 0.58% per
greater than 2 on the Michigan Neuropathy year (95% CI –0.01 to 1.17) [73]. While glycemic
Screening Instrument (MNSI). 1277 of 2815 par- control did not demonstrate a statistically signifi-
ticipants in the intensive arm and 1338 of 2791 in cant decrease in the incidence of neuropathy in
conventional arm developed neuropathy. Overall any of these four studies or in the meta-analysis,
these findings demonstrated a 0.70% per year the consistent findings indicate a small, but real
risk reduction (95% CI –0.01 to 1.40) and a non-­ effect of glycemic control. However, in compari-
significant 5% relative risk reduction in those son to the studies in type 1 diabetes, the effect of
receiving intensive therapy [93]. glycemic control is much less. These results high-
The second largest study was Duckworth light the importance of factors other than hyper-
2009, which investigated 1791 military veterans glycemia in the development of neuropathy in type
who had a suboptimal response to therapy for 2 patients. Furthermore, glycemic control is essen-
type 2 diabetes and randomized them to receive tial in patients with type 1 diabetes.
either intensive or standard glucose control for a While not the primary outcome of this Cochrane
median of 5.6 years. Participants in the intensive review, the largest study that reported neuropathy
therapy group were started on maximal doses, outcomes in patients with type 2 patients was the
whereas those in the standard therapy group were UKPDS Study Group 1998, which followed 3867
started on half the maximal doses. Clinical neu- individuals with newly diagnosed type 2 diabetes
ropathy in this study was defined as a diagnosis who were randomly assigned to an intensive arm
Neuropathy in Type 1 and Type 2 Diabetes 61

with a sulphonylurea (chlorpropamide, gliben- which included increased weight gain (RR 1.96,
clamide, or glipizide) or insulin, or to a conven- 95% CI 1.81–2.13) and deaths (RR 1.26, 95% CI
tional policy with diet. Neuropathy was defined as 1.06–1.51) in those in the intensive group [93].
a vibration threshold greater than 25 V on a bioth- Since the 2012 Cochrane review, only one
esiometer. Participants were followed for as many study has evaluated the effect of glycemic control
as 15 years. They found that there was a modest on neuropathy and this was in patients with type 2
risk reduction in favor of intensive treatment, but diabetes. Specifically, the ADDITION (Anglo-­
this result was only statistically significant at 15 Danish-­ Dutch Study of Intensive Treatment in
years [94]. Shichiri et al. [124] investigated nerve People With Screen Detected Diabetes in Primary
conduction study findings in a randomized trial of Care)-Europe study evaluated if intensive treat-
intensive glycemic control compared to conven- ment (IT) improved outcomes compared with
tional teratment in patients with type 2 diabetes. A routine care (RC). This trial comprised of two
total of 110 participants were followed for 8 years phases, a screening phase and a randomization
with type 2 diabetes (55 with no retinopathy—pri- phase involving 343 general practices throughout
mary prevention cohort, and 55 with simple reti- Denmark; Cambridge, UK; the Netherlands; and
nopathy—secondary intervention cohort) and Leicester, UK. Participants were included if they
assigned to intensive insulin therapy (three or were 40–69 years (50–69 years in the Netherlands)
more daily injections) or to conventional insulin without known diabetes. Screening comprised of
therapy (one to two daily injections). There was an various measures depending on what country the
increase of 0.44 (0.09) m/s in the intensive arm patient was located in, and included oral glucose
compared to a decline of 0.13 (0.08) m/s in the tolerance tests, or random glucose measurements
conventional arm with a statistically significant and HgbA1c followed by fasting glucose and oral
annual MD of 0.56 m/s (95% CI 0.53–0.60) in glucose tolerance test. All patients with newly
favor of the intensive group [124]. Interestingly, diagnosed type 2 diabetes mellitus were eligible
there was a similar effect in conduction velocity to participate and randomized between IT and RC
changes seen in type 1 diabetes, even though there in a 1:1 ratio. Ultimately, 3057 participants took
was a much smaller relative risk reduction of clini- part in this trial and 196 participants died before
cal neuropathy in participants with type 2 diabetes follow-up. Questionnaire data on peripheral neu-
compared to type 1 diabetes. These results support ropathy was collected in 2312 participants.
the importance of glycemic control in patients Targets for the IT arm were HgbA1c less than
with type 2 diabetes even if it is not as important as 7.0%, blood pressure less than or equal to 135/85,
it is in type 1 diabetes. cholesterol less than 5 mmol/L in patients without
Similar to the trials evaluating patients with ischemic heart disease or less than 4.5 mmol/L in
type 1 diabetes, those with type 2 diabetes also patients with ischemic heart disease. Treatments
suffered more adverse of events in the intensive were recommended but therapy decisions were
group in the three largest studies with the longest ultimately deferred to the prescribing clinicians
follow-up. Specifically, there was an approxi- and patients. After 5.3 years, 1296 subjects in the
mately threefold higher risk of severe hypoglyce- IT arm, and 1016 subjects in the RC arm were
mic episodes in those receiving intensive therapy. evaluated. Ultimately 4.9% participants in the IT
In contrast to participants with type 1 diabetes, arm screened positive for peripheral neuropathy
the effect of intensive glycemic control and neu- based on the MNSI questionnaire compared to
ropathy was much less impressive, and in this 5.9% of participants in the RC group [125].
group the risk of hypoglycemia still remains sub- Therefore, no significant difference was seen in
stantial so this risk needs to be taken into account decreasing the incidence of neuropathy in this
when determining the risk versus benefit ratio of population of patients with screen detected type 2
enhanced glucose control [73]. Importantly, the diabetes with intensive therapy. These results are
ACCORD study demonstrated an increase in very similar to the previous four studies in patients
mortality when targeting a HbA1C of less than with type 2 diabetes described above. As a result,
6%, which highlights the importance of tradeoffs the 2012 Cochrane review conclusions remain
between glycemic control and adverse events, relevant. Namely that glycemic control is more
62 G. Akinci et al.

important in patients with type 1 diabetes com- intensive glycemic control has a much smaller
pared with patients with type 2 diabetes. effect on preventing neuropathy in patients with
While many studies of glycemic control have type 2 diabetes, which highlights the need for
been performed to determine its effect on neuropa- novel therapies addressing other metabolic factors
thy outcomes, few studies have addressed the in this population. Based on the LOOK Ahead
effects of weight loss on neuropathy outcomes. study, weight loss is a promising intervention, but
The main study addressing this intervention is the future studies are needed to verify this finding.
Look Ahead trial, which randomized 5145 over-
weight or obese people with type 2 diabetes to
intensive lifestyle intervention (ILI) (n = 2570) or References
to a diabetes support and education (DSE) control 1. American Diabetes Association American Academy
group (n = 2575). Staff responsible for obtaining of Neurology. Consensus statement: report and rec-
data on study outcomes were blinded, but because ommendations of the San Antonio conference on dia-
of the types of interventions being evaluated, betic neuropathy. Diabetes Care. 1988;11(7):592–7.
2. Dyck PJ, Albers JW, Andersen H, et al. Diabetic poly-
patients and interventionists were not blinded. neuropathies: update on research definition, diagnos-
Interventions were terminated 9–11 years after tic criteria and estimation of severity. Diabetes Metab
randomization, but both groups continued to be Res Rev. 2011;27(7):620–8.
followed for primary and secondary outcomes. 3. Feldman EL, Callaghan BC, Pop-Busui R, et  al.
Diabetic neuropathy. Nat Rev Dis Primers.
Neuropathy evaluations included the MNSI ques- 2019;5(1):41.
tionnaire completed at baseline in all participants 4. Morrison S, Colberg SR, Parson HK, Vinik AI. Relation
and repeated annually, and the MNSI examination. between risk of falling and postural sway complexity
The baseline MNSI questionnaire scores were 1.9 in diabetes. Gait Posture. 2012;35(4):662–8.
5. Author. Report and recommendations of the
± 0.04 and 1.8 ± 0.04 in the ILI and DSE groups, San Antonio conference on diabetic neuropathy.
respectively, which was not statistically signifi- Neurology. 1988;38(7):1161–5.
cant. After 1 year when weight loss was maximal, 6. Pop-Busui R, Boulton AJ, Feldman EL, et  al.
the ILI group had a statistically significant lower Diabetic neuropathy: a position statement by the
American Diabetes Association. Diabetes Care.
MNSI questionnaire score compared to the DSE 2017;40(1):136–54.
group (1.7 ± 0.04 and 2.0 ± 0.04, p < 0.001). There 7. Visser NA, Notermans NC, Linssen RS, van den
was also a significant association between changes Berg LH, Vrancken AF. Incidence of polyneu-
in the MNSI questionnaire scores and changes in ropathy in Utrecht, the Netherlands. Neurology.
2015;84(3):259-264.
body weight, HgbA1c, and serum lipids. However, 8. Forrest KY, Maser RE, Pambianco G, Becker DJ,
there was no significant between-group differ- Orchard TJ.  Hypertension as a risk factor for dia-
ences in the proportion of participants with MNSI betic neuropathy: a prospective study. Diabetes.
examination scores greater than or equal to 2.5. 1997;46(4):665–70.
9. Sands ML, Shetterly SM, Franklin GM, Hamman
Overall, this study demonstrated that ILI resulted RF. Incidence of distal symmetric (sensory) neuropa-
in a significant decrease in questionnaire based, thy in NIDDM: the San Luis Valley Diabetes Study.
but not examination based neuropathy. Therefore, Diabetes Care. 1997;20(3):322-329.
weight loss likely reduces neuropathy in those 10. Jenkins PJ, Srikantharajah D, Duckworth AD, Watts
AC, McEachan JE. Carpal tunnel syndrome: the asso-
patients with type 2 diabetes that are also over- ciation with occupation at a population level. J Hand
weight or obese. However, the magnitude of effect Surg Eur Vol. 2013;38(1):67-72.
is likely small highlighting the need for novel 11. Latinovic R, Gulliford MC, Hughes RA. Incidence of
treatments to prevent neuropathy. common compressive neuropathies in primary care. J
Neurol Neurosurg Psychiatry. 2006;77(2): 263-265.
In conclusion, studies comparing different lev- 12. Falkiner S, Myers S. When exactly can carpal tunnel
els of glycemic control on the development of syndrome be considered work-related? ANZ J Surg.
neuropathy in patients with diabetes reveal clear 2002;72(3):204-209.
differences between type 1 and type 2 diabetes. 13. Mondelli M, Fiannini F, Mondelli M, Giannini F,
BalleriniM, Ginanneschi F, Martorelli E. Incidence of
Intensive glycemic control reduces the develop- ulner neuropathy at the elbow in the province of Siena
ment of neuropathy in patients with type 1 diabe- (Italy). J Neurol Sci. 2005;234:5-10.
tes, which emphasizes the importance of 14. van Slobbe AM, Bohnen AM, Bernsen RM, Koes
hyperglycemia in this population. In contrast, BW, Bierma-Zeinstra SM. Incidence rates and deter-
Neuropathy in Type 1 and Type 2 Diabetes 63

minants in meralgia paresthetica in general practice. J 31. Skyler JS, Bakris GL, Bonifacio E, et al. Differentiation
Neurol. 2004;251(3):294-297. of diabetes by pathophysiology, natural history, and
15. Rowlands S, Hooper R, Hughes R, Burney P. The epi- prognosis. Diabetes. 2017;66(2):241–55.
demiology and treatment of Bell’s palsy in the UK. 32. Unger RH, Grundy S. Hyperglycaemia as an inducer
Eur J Neurol. 2002;9(1):63-67. as well as a consequence of impaired islet cell function
16. Katusic SK, Beard CM, WiederholtWC, Bergstralh and insulin resistance: implications for the manage-
EJ, Kurland LT. Incidence, clinical features, and prog- ment of diabetes. Diabetologia. 1985;28(3):119–21.
nosis in Bell’s palsy, Rochester, Minnesota, 1968- 33. Partanen J, Niskanen L, Lehtinen J, Mervaala E,
1982. Ann Neurol. 1986;20(5): 622-627. Siitonen O, Uusitupa M. Natural history of peripheral
17. Monini S, Lazzarino AI, Iacolucci C, Buffoni A, neuropathy in patients with non-­ insulin-dependent
BarbaraM. Epidemiology of Bell’s palsy in an Italian diabetes mellitus. N Engl J Med. 1995;333(2):89–94.
Health District: incidence and case-control study. 34. Sumner CJ, Sheth S, Griffin JW, Cornblath DR,
Acta Otorhinolaryngol Ital. 2010;30(4):198. Polydefkis M.  The spectrum of neuropathy in dia-
18. Schoenfeld AJ, Laughlin M, Bader JO, Bono CM. betes and impaired glucose tolerance. Neurology.
Characterization of the incidence and risk factors for 2003;60(1):108–11.
the development of lumbar radiculopathy. J Spinal 35. Singleton JR, Smith AG, Bromberg MB. Painful sen-
Disord Tech. 2012;25(3):163-167. sory polyneuropathy associated with impaired glu-
19. Schoenfeld AJ, George AA, Bader JO, Caram PM Jr. cose tolerance. Muscle Nerve. 2001;24(9):1225–8.
Incidence and epidemiology of cervical radiculopathy 36. Sveen KA, Karime B, Jorum E, et  al. Small- and
in the United Statesmilitary: 2000 to 2009. J Spinal large-fiber neuropathy after 40 years of type 1 diabe-
Disord Tech. 2012;25(1):17-22. tes: associations with glycemic control and advanced
20. Radhakrishnan K, LitchyWJ, O’Fallon WM, Kurland protein glycation: the Oslo Study. Diabetes Care.
LT. Epidemiology of cervical radiculopathy: a popu- 2013;36(11):3712–7.
lation-based study from Rochester, Minnesota, 1976 37. Alam U, Jeziorska M, Petropoulos IN, et  al. Latent
through 1990. Brain. 1994;117(pt 2):325-335. autoimmune diabetes of adulthood (LADA) is asso-
21. Tracy JA, Dyck PJ.  The spectrum of diabetic ciated with small fibre neuropathy. Diabet Med.
neuropathies. Phys Med Rehabil Clin N Am. 2019;36(9):1118–24.
2008;19(1):1–26. 38. Dyck PJ, Kratz KM, Karnes JL, et al. The prevalence
22. Breiner A, Lovblom LE, Perkins BA, Bril V. Does by staged severity of various types of diabetic neu-
the prevailing hypothesis that small-fiber dys- ropathy, retinopathy, and nephropathy in a popula-
function precedes large-fiber dysfunction apply tion-based cohort: the Rochester Diabetic Neuropathy
to type 1 diabetic patients? Diabetes Care. Study. Neurology. 1993;43(4):817–24.
2014;37(5):1418–24. 39. Loseth S, Stalberg EV, Lindal S, Olsen E, Jorde R,
23. Feldman EL, Nave KA, Jensen TS, Bennett DLH. New Mellgren SI. Small and large fiber neuropathy in those
horizons in diabetic neuropathy: mechanisms, bioen- with type 1 and type 2 diabetes: a 5-year follow-­up
ergetics, and pain. Neuron. 2017;93(6):1296–313. study. J Peripher Nerv Syst. 2016;21(1):15–21.
24. American Diabetes A.  Microvascular complications 40. Loseth S, Mellgren SI, Jorde R, Lindal S, Stalberg
and foot care: standards of medical care in diabetes- E.  Polyneuropathy in type 1 and type 2 diabetes:
­2021. Diabetes Care. 2021;44(1):151–67. comparison of nerve conduction studies, thermal per-
25. Abbott CA, Malik RA, van Ross ER, Kulkarni J, ception thresholds and intraepidermal nerve fibre den-
Boulton AJ.  Prevalence and characteristics of pain- sities. Diabetes Metab Res Rev. 2010;26(2):100–6.
ful diabetic neuropathy in a large community-­based 41. Vinik AI, Casellini C, Parson HK, Colberg SR,
diabetic population in the UK.  Diabetes Care. Nevoret ML.  Cardiac autonomic neuropathy in dia-
2011;34(10):2220–4. betes: a predictor of cardiometabolic events. Front
26. Didangelos T, Doupis J, Veves A.  Painful diabetic Neurosci. 2018;12:591.
neuropathy: clinical aspects. Handb Clin Neurol. 42. Maser RE, Mitchell BD, Vinik AI, Freeman R. The
2014;126:53–61. association between cardiovascular autonomic neu-
27. Boulton AJ, Armstrong WD, Scarpello JH, Ward ropathy and mortality in individuals with diabetes: a
JD. The natural history of painful diabetic neuropathy– meta-analysis. Diabetes Care. 2003;26(6):1895–901.
a 4-year study. Postgrad Med J. 1983;59(695):556–9. 43. Ziegler D, Zentai CP, Perz S, et al. Prediction of mor-
28. Benbow SJ, Chan AW, Bowsher D, MacFarlane IA, tality using measures of cardiac autonomic dysfunc-
Williams G.  A prospective study of painful symp- tion in the diabetic and nondiabetic population: the
toms, small-fibre function and peripheral vascular MONICA/KORA Augsburg Cohort Study. Diabetes
disease in chronic painful diabetic neuropathy. Diabet Care. 2008;31(3):556–61.
Med. 1994;11(1):17–21. 44. Pfeifer MA, Weinberg CR, Cook DL, et al. Autonomic
29. Katoulis EC, Ebdon-Parry M, Lanshammar H, neural dysfunction in recently diagnosed diabetic sub-
Vileikyte L, Kulkarni J, Boulton AJ.  Gait abnor- jects. Diabetes Care. 1984;7(5):447–53.
malities in diabetic neuropathy. Diabetes Care. 45. Jaiswal M, Divers J, Urbina EM, et al. Cardiovascular
1997;20(12):1904–7. autonomic neuropathy in adolescents and young
30. Walters DP, Gatling W, Mullee MA, Hill RD.  The adults with type 1 and type 2 diabetes: The SEARCH
prevalence of diabetic distal sensory neuropathy in an for Diabetes in Youth Cohort Study. Pediatr Diabetes.
English community. Diabet Med. 1992;9(4):349–53. 2018;19(4):680–9.
64 G. Akinci et al.

46. Eleftheriadou A, Williams S, Nevitt S, et al. The preva- 62. Dyck PJ, Windebank AJ.  Diabetic and nondiabetic
lence of cardiac autonomic neuropathy in prediabetes: a lumbosacral radiculoplexus neuropathies: new
systematic review. Diabetologia. 2021;64(2):288–303. insights into pathophysiology and treatment. Muscle
47. Martin CL, Albers JW, Pop Busui R. Neuropathy and Nerve. 2002;25(4):477–91.
related findings in the diabetes control and complica- 63. Boulton AJ, Malik RA, Arezzo JC, Sosenko
tions trial/epidemiology of diabetes interventions and JM.  Diabetic somatic neuropathies. Diabetes Care.
complications study. Diabetes Care. 2014;37(1):31–8. 2004;27(6):1458–86.
48. Pop-Busui R, Low PA, Waberski BH, et  al. Effects 64. Krol CG, Klok FA, de Koning EJ. Diplopia as the pre-
of prior intensive insulin therapy on cardiac auto- senting symptom of type 1 diabetes. Diabetes Care.
nomic nervous system function in type 1 diabetes 2014;37(3):e45–6.
mellitus: the Diabetes Control and Complications 65. Keane JR.  Third nerve palsy: analysis of 1400 per-
Trial/Epidemiology of Diabetes Interventions and sonally-examined inpatients. Can J Neurol Sci.
Complications study (DCCT/EDIC). Circulation. 2010;37(5):662–70.
2009;119(22):2886–93. 66. Chou KL, Galetta SL, Liu GT, et  al. Acute ocular
49. Choung RS, Locke GR, Schleck CD, Zinsmeister AR, motor mononeuropathies: prospective study of the
Melton LJ, Talley NJ. Risk of gastroparesis in subjects roles of neuroimaging and clinical assessment. J
with type 1 and 2 diabetes in the general population. Neurol Sci. 2004;219(1-2):35–9.
Am J Gastroenterol. 2012;107(1):82–8. 67. Vinik A, Mehrabyan A, Colen L, Boulton A.  Focal
50. Krishnasamy S, Abell TL.  Diabetic gastroparesis: entrapment neuropathies in diabetes. Diabetes Care.
principles and current trends in management. Diabetes 2004;27(7):1783–8.
Ther. 2018;9(Suppl 1):1–42. 68. Ziegler D, Papanas N, Vinik AI, Shaw
51. Freeman R.  Diabetic autonomic neuropathy. Handb JE. Epidemiology of polyneuropathy in diabetes and
Clin Neurol. 2014;126:63–79. prediabetes. Handb Clin Neurol. 2014;126:3–22.
52. Valdovinos MA, Camilleri M, Zimmerman 69. Tesfaye S, Stevens LK, Stephenson JM, et  al.
BR.  Chronic diarrhea in diabetes mellitus: mecha- Prevalence of diabetic peripheral neuropathy and its
nisms and an approach to diagnosis and treatment. relation to glycaemic control and potential risk fac-
Mayo Clin Proc. 1993;68(7):691–702. tors: the EURODIAB IDDM Complications Study.
53. Lysy J, Israeli E, Goldin E. The prevalence of chronic Diabetologia. 1996;39(11):1377–84.
diarrhea among diabetic patients. Am J Gastroenterol. 70. Adler AI, Boyko EJ, Ahroni JH, Stensel V, Forsberg
1999;94(8):2165–70. RC, Smith DG.  Risk factors for diabetic periph-
54. Kempler P, Amarenco G, Freeman R, et al. Management eral sensory neuropathy. Results of the seattle
strategies for gastrointestinal, erectile, bladder, and prospective diabetic foot study. Diabetes Care.
sudomotor dysfunction in patients with diabetes. 1997;20(7):1162–7.
Diabetes Metab Res Rev. 2011;27(7):665–77. 71. Braffett BH, Gubitosi-Klug RA, Albers JW, et al. Risk
55. Araujo AB, Hall SA, Ganz P, et al. Does erectile dys- factors for diabetic peripheral neuropathy and cardio-
function contribute to cardiovascular disease risk pre- vascular autonomic neuropathy in the diabetes con-
diction beyond the Framingham risk score? J Am Coll trol and complications trial/epidemiology of diabetes
Cardiol. 2010;55(4):350–6. interventions and complications (DCCT/EDIC) study.
56. Pop-Busui R, Hotaling J, Braffett BH, et  al. Diabetes. 2020;69(5):1000–10.
Cardiovascular autonomic neuropathy, erectile dys- 72. Callaghan BC, Gallagher G, Fridman V, Feldman
function and lower urinary tract symptoms in men EL. Diabetic neuropathy: what does the future hold?
with type 1 diabetes: findings from the DCCT/EDIC. J Diabetologia. 2020;63(5):891–7.
Urol. 2015;193(6):2045–51. 73. Callaghan BC, Little AA, Feldman EL, Hughes
57. Garcia-Cruz E, Leibar-Tamayo A, Romero J, et  al. RA.  Enhanced glucose control for preventing and
Metabolic syndrome in men with low testosterone treating diabetic neuropathy. Cochrane Database Syst
levels: relationship with cardiovascular risk factors Rev. 2012;6:CD007543.
and comorbidities and with erectile dysfunction. J Sex 74. Ziegler D, Behler M, Schroers-Teuber M, Roden
Med. 2013;10(10):2529–38. M.  Near-normoglycaemia and development of neu-
58. Sarma AV, Kanaya A, Nyberg LM, et al. Risk factors ropathy: a 24-year prospective study from diagnosis
for urinary incontinence among women with type 1 of type 1 diabetes. BMJ Open. 2015;5(6):e006559.
diabetes: findings from the epidemiology of diabe- 75. Maser RE, Steenkiste AR, Dorman JS, et  al.
tes interventions and complications study. Urology. Epidemiological correlates of diabetic neuropathy.
2009;73(6):1203–9. Report from Pittsburgh Epidemiology of Diabetes
59. Albers JW, Pop-Busui R. Diabetic neuropathy: mech- Complications Study. Diabetes. 1989;38(11):1456–61.
anisms, emerging treatments, and subtypes. Curr 76. Callaghan BC, Hur J, Feldman EL.  Diabetic neu-
Neurol Neurosci Rep. 2014;14(8):473. ropathy: one disease or two? Curr Opin Neurol.
60. Gibbons CH, Freeman R. Treatment-induced neurop- 2012;25(5):536–41.
athy of diabetes: an acute, iatrogenic complication of 77. Pop-Busui R, Braffett BH, Zinman B, et  al.
diabetes. Brain. 2015;138(Pt 1):43–52. Cardiovascular autonomic neuropathy and car-
61. Gibbons CH. Treatment induced neuropathy of diabe- diovascular outcomes in the diabetes control and
tes. Auton Neurosci. 2020;226:102668. complications trial/epidemiology of diabetes inter-
Neuropathy in Type 1 and Type 2 Diabetes 65

ventions and complications (DCCT/EDIC) study. 92. Ohkubo Y, Kishikawa H, Araki E, et  al. Intensive
Diabetes Care. 2017;40(1):94–100. insulin therapy prevents the progression of diabetic
78. Pambianco G, Costacou T, Ellis D, Becker DJ, microvascular complications in Japanese patients
Klein R, Orchard TJ.  The 30-year natural history with non-insulin-dependent diabetes mellitus: a ran-
of type 1 diabetes complications: the Pittsburgh domized prospective 6-year study. Diabetes Res Clin
Epidemiology of Diabetes Complications Study Pract. 1995;28(2):103–17.
experience. Diabetes. 2006;55(5):1463–9. 93. Ismail-Beigi F, Craven T, Banerji MA, et  al.
79. Tesfaye S, Chaturvedi N, Eaton SE, et al. Vascular Effect of intensive treatment of hyperglycaemia
risk factors and diabetic neuropathy. N Engl J Med. on microvascular outcomes in type 2 diabetes: an
2005;352(4):341–50. analysis of the ACCORD randomised trial. Lancet.
80. Jaiswal M, Divers J, Dabelea D, et  al. Prevalence 2010;376(9739):419–30.
of and risk factors for diabetic peripheral neu- 94. UK Prospective Diabetes Study (UKPDS) Group.
ropathy in youth with type 1 and type 2 diabetes: Intensive blood-glucose control with sulpho-
SEARCH for Diabetes in Youth Study. Diabetes nylureas or insulin compared with conventional
Care. 2017;40(9):1226–32. treatment and risk of complications in patients
81. Akinci G, Savelieff MG, Gallagher G, Callaghan with type 2 diabetes (UKPDS 33). Lancet.
BC, Feldman EL.  Diabetic neuropathy in children 1998;352(9131):837–53.
and youth: new and emerging risk factors. Pediatr 95. Duckworth W, Abraira C, Moritz T, et  al.
Diabetes. 2020;22(2):132–47. Glucose control and vascular complications in
82. Singleton JR, Smith AG, Bromberg MB. Increased veterans with type 2 diabetes. N Engl J Med.
prevalence of impaired glucose tolerance in patients 2009;360(2):129–39.
with painful sensory neuropathy. Diabetes Care. 96. Charles M, Ejskjaer N, Witte DR, Borch-Johnsen
2001;24(8):1448–53. K, Lauritzen T, Sandbaek A. Prevalence of neuropa-
83. Asghar O, Petropoulos IN, Alam U, et  al. Corneal thy and peripheral arterial disease and the impact
confocal microscopy detects neuropathy in subjects of treatment in people with screen-detected type 2
with impaired glucose tolerance. Diabetes Care. diabetes: the ADDITION-Denmark study. Diabetes
2014;37(9):2643–6. Care. 2011;34(10):2244–9.
84. Bongaerts BW, Rathmann W, Heier M, et al. Older 97. Charles M, Fleischer J, Witte DR, et  al. Impact
subjects with diabetes and prediabetes are fre- of early detection and treatment of diabetes on
quently unaware of having distal sensorimotor poly- the 6-year prevalence of cardiac autonomic neu-
neuropathy: the KORA F4 study. Diabetes Care. ropathy in people with screen-detected diabetes:
2013;36(5):1141–6. ADDITION-Denmark, a cluster-randomised study.
85. Callaghan BC, Xia R, Banerjee M, et al. Metabolic Diabetologia. 2013;56(1):101–8.
syndrome components are associated with symp- 98. Siegelaar SE, Kilpatrick ES, Rigby AS, Atkin SL,
tomatic polyneuropathy independent of glycemic Hoekstra JB, Devries JH.  Glucose variability does
status. Diabetes Care. 2016;39(5):801–7. not contribute to the development of peripheral and
86. Callaghan BC, Reynolds EL, Banerjee M, et al. The autonomic neuropathy in type 1 diabetes: data from
prevalence and determinants of cognitive deficits the DCCT. Diabetologia. 2009;52(10):2229–32.
and traditional diabetic complications in the severely 99. Xu F, Zhao LH, Su JB, et  al. The relationship
obese. Diabetes Care. 2020;43(3):683–90. between glycemic variability and diabetic peripheral
87. Callaghan BC, Reynolds E, Banerjee M, Chant E, neuropathy in type 2 diabetes with well-controlled
Villegas-Umana E, Feldman EL. Central obesity is HbA1c. Diabetol Metab Syndr. 2014;6(1):139.
associated with neuropathy in the severely obese. 100. Kwai NC, Arnold R, Poynten AM, Krishnan
Mayo Clin Proc. 2020;95(7):1342–53. AV.  Association between glycemic variability and
88. Callaghan BC, Xia R, Reynolds E, et al. Association peripheral nerve dysfunction in type 1 diabetes.
between metabolic syndrome components and poly- Muscle Nerve. 2016;54(5):967–9.
neuropathy in an obese population. JAMA Neurol. 101. Lachin JM, Bebu I, Bergenstal RM, et al. Association
2016;73(12):1468–76. of glycemic variability in type 1 diabetes with pro-
89. Callaghan BC, Gao L, Li Y, et al. Diabetes and obesity gression of microvascular outcomes in the diabe-
are the main metabolic drivers of peripheral neurop- tes control and complications trial. Diabetes Care.
athy. Ann Clin Transl Neurol. 2018;5(4):397–405. 2017;40(6):777–83.
90. Schlesinger S, Herder C, Kannenberg JM, et  al. 102. Christensen MMB, Hommel EE, Jorgensen ME,
General and abdominal obesity and incident dis- Fleischer J, Hansen CS.  Glycemic variability and
tal sensorimotor polyneuropathy: insights into diabetic neuropathy in young adults with type 1 dia-
inflammatory biomarkers as potential media- betes. Front Endocrinol. 2020;11:644.
tors in the KORA F4/FF4 cohort. Diabetes Care. 103. Ortega FB, Lavie CJ, Blair SN. Obesity and cardio-
2019;42(2):240–7. vascular disease. Circ Res. 2016;118(11):1752–70.
91. Hanewinckel R, Drenthen J, Ligthart S, et  al. 104. Andersen ST, Witte DR, Dalsgaard EM, et al. Risk
Metabolic syndrome is related to polyneuropa- factors for incident diabetic polyneuropathy in a
thy and impaired peripheral nerve function: a pro- cohort with screen-detected type 2 diabetes followed
spective population-based cohort study. J Neurol for 13 years: ADDITION-denmark. Diabetes Care.
Neurosurg Psychiatry. 2016;87(12):1336–42. 2018;41(5):1068–75.
66 G. Akinci et al.

105. Callaghan BC, Price RS, Feldman EL. Distal symmet- 116. Callaghan BC, Feldman E, Liu J, et al. Triglycerides
ric polyneuropathy in 2020. JAMA. 2020;324(1):90–1. and amputation risk in patients with diabetes: ten-­
106. Shah AS, El Ghormli L, Vajravelu ME, et al. Heart year follow-up in the DISTANCE study. Diabetes
rate variability and cardiac autonomic dysfunction: Care. 2011;34(3):635–40.
prevalence, risk factors, and relationship to arterial 117. Andersen ST, Witte DR, Fleischer J, et al. Risk factors
stiffness in the treatment options for type 2 diabetes for the presence and progression of cardiovascular
in adolescents and youth (TODAY) study. Diabetes autonomic neuropathy in type 2 diabetes: ADDITION-
Care. 2019;42(11):2143–50. Denmark. Diabetes Care. 2018;41(12):2586–94.
107. Laitinen T, Lindstrom J, Eriksson J, et  al. 118. Author. Effect of intensive diabetes treatment on nerve
Cardiovascular autonomic dysfunction is associated conduction in the Diabetes Control and Complications
with central obesity in persons with impaired glu- Trial. Ann Neurol. 1995;38(6):869–80.
cose tolerance. Diabet Med. 2011;28(6):699–704. 119. Diabetes C, Nathan DM, et al. The effect of inten-
108. Dabelea D, Stafford JM, Mayer-Davis EJ, et  al. sive treatment of diabetes on the development
Association of type 1 diabetes vs type 2 diabetes and progression of long-term complications in
diagnosed during childhood and adolescence with insulin-­dependent diabetes mellitus. N Engl J Med.
complications during teenage years and young adult- 1993;329(14):977–86.
hood. JAMA. 2017;317(8):825–35. 120. Ponirakis G, Petropoulos IN, Alam U, et  al.
109. De Block CE, De Leeuw IH, Van Gaal LF. Impact Hypertension contributes to neuropathy in patients with
of overweight on chronic microvascular complica- type 1 diabetes. Am J Hypertens. 2019;32(8):796–803.
tions in type 1 diabetic patients. Diabetes Care. 121. Linn T, Ortac K, Laube H, Federlin K.  Intensive
2005;28(7):1649–55. therapy in adult insulin-dependent diabetes mel-
110. Wiggin TD, Sullivan KA, Pop-Busui R, Amato A, litus is associated with improved insulin sensitivity
Sima AA, Feldman EL.  Elevated triglycerides cor- and reserve: a randomized, controlled, prospective
relate with progression of diabetic neuropathy. study over 5 years in newly diagnosed patients.
Diabetes. 2009;58(7):1634–40. Metabolism. 1996;45(12):1508–13.
111. Han L, Ji L, Chang J, et al. Peripheral neuropathy is 122. Azad N, Emanuele NV, Abraira C, et  al. The
associated with insulin resistance independent of met- effects of intensive glycemic control on neuropa-
abolic syndrome. Diabetol Metab Syndr. 2015;7:14. thy in the VA cooperative study on type II diabe-
112. Ylitalo KR, Sowers M, Heeringa S. Peripheral vas- tes mellitus (VA CSDM). J Diabetes Complicat.
cular disease and peripheral neuropathy in individu- 1999;13(5-6):307–13.
als with cardiometabolic clustering and obesity: 123. Tovi J, Svanborg E, Nilsson BY, Engfeldt P. Diabetic
National Health and Nutrition Examination Survey neuropathy in elderly Type 2 diabetic patients:
2001-2004. Diabetes Care. 2011;34(7):1642–7. effects of insulin treatment. Acta Neurol Scand.
113. Smith AG, Singleton JR.  Obesity and hyperlipid- 1998;98(5):346–53.
emia are risk factors for early diabetic neuropathy. J 124. Shichiri M, Kishikawa H, Ohkubo Y, Wake N. Long-­
Diabetes Complicat. 2013;27(5):436–42. term results of the Kumamoto Study on optimal
114. Lee KO, Nam JS, Ahn CW, et al. Insulin resistance is diabetes control in type 2 diabetic patients. Diabetes
independently associated with peripheral and auto- Care. 2000;23(2):21–9.
nomic neuropathy in Korean type 2 diabetic patients. 125. Sandbaek A, Griffin SJ, Sharp SJ, et  al. Effect of
Acta Diabetol. 2012;49(2):97–103. early multifactorial therapy compared with routine
115. Cai Z, Yang Y, Zhang J.  A systematic review and care on microvascular outcomes at 5 years in people
meta-analysis of the serum lipid profile in prediction with screen-detected diabetes: a randomized con-
of diabetic neuropathy. Sci Rep. 2021;11(1):499. trolled trial: the ADDITION-Europe Study. Diabetes
Care. 2014;37(7):2015–23.
Clinical Diagnosis of Diabetic
Peripheral Neuropathy

Bruce A. Perkins and Vera Bril

1 Introduction and Context agement, consider other causes of polyneuropa-


thy, while identifying atypical features that
The variability in clinical presentation and com- warrant specialist referral. This chapter discusses
plexities in measurement of diabetic neuropathy these considerations and the approaches to the
have not only challenged the clear understanding clinical diagnosis of diabetic neuropathy, while
of epidemiology, impact, and the clinical trial chapter “Clinical Features Diabetic Neuropathies”
development of therapies, but these often compli- details clinical features, and chapter “Diagnostic
cate the diagnostic process. However, with a clear Techniques for Diabetic Peripheral Neuropathy”
understanding of these obstacles and the relevant details specialized diagnostic techniques.
clinical tools, clinicians from all fields of spe-
cialty can develop an efficient approach to diag-
nosis. This includes more simplified approaches 1.1 Diabetic Distal Symmetric
suitable to primary care and diabetes management Polyneuropathy (Diabetic
clinics that screen for and diagnose diabetic neu- DSP): A Brief Overview
ropathy, to the specialty neurology clinics that
may use more refined diagnostic methods required The most common form—and the one usually
for confirmation of neuropathy and the evaluation referred to as “diabetic peripheral neuropathy” or
of more atypical cases. Certainly, the vast major- even more simply as “diabetic neuropathy”—is
ity of cases of diabetic neuropathy do not require the diffuse, symmetrical, slowly-progressive
referral to a neurology specialist for confirmation. length-dependent damage to the peripheral and
This places a responsibility on the diabetes care autonomic nervous system classified technically
clinician to be able to confidently make a working by the term “diabetic distal symmetrical polyneu-
diagnosis on clinical evidence and to initiate man- ropathy” (diabetic DSP) [1]. It typically remains
asymptomatic for years, may first present clini-
cally with symptoms of abnormal sensation sym-
B. A. Perkins (*) metrically at the tips of the toes and may over
Division of Endocrinology, Department of Medicine,
Sinai Health System, University of Toronto, time spread to the stocking-and-glove distribu-
Toronto, ON, Canada tion. It involves injury to different anatomical
e-mail: Bruce.Perkins@sinaihealth.ca nerve types that show variable clinical manifesta-
V. Bril tions between individuals. These can be classi-
Division of Neurology, Department of Medicine, fied as small fibers (thinly-myelinated autonomic
University of Toronto, Toronto, ON, Canada B-fibers, and thinly- or un- myelinated autonomic
e-mail: vera.bril@utoronto.ca

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 67


S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_5
68 B. A. Perkins and V. Bril

and sensory C-fibers) that make up autonomic the subsequent risk of ulcer, infection, and ampu-
and pain and temperature sensory fibers. Damage tation intensify. Complications like these are
to large fibers (myelinated Aα, Aβ, Aδ sensory, feared more than death itself [4], and of great
and Aγ motor) are responsible for other sensory concern is the recent evidence in certain parts of
and skeletal muscle impairments [2, 3]. At the the world of a resurgence in the risk of amputa-
symptomatic stages, some have a large fiber-­ tions [5–7].
predominant pattern and experience numbness,
tingling, or imbalance owing to sensory ataxia,
while others have small fiber-predominant symp- 1.2 Staging of Diabetic DSP
toms that may instead present with burning and
stabbing pain, impairment in sensing heat and For the purpose of understanding the face value
cold, or a propensity to clinical autonomic abnor- of symptoms and signs and their measurement
malities like postural hypotension and gastropa- scales, we center the discussion of clinical diag-
resis [1]. Some experience combined nosis in this chapter around the conventional
manifestations of large and small fiber types, and, staging system described in Table 1 [9]. In simple
regardless of pattern, people with diabetes can be terms, in practice diabetic DSP (Stage 1b or
asymptomatic for extended periods even though higher) is identified through two possible points
their physical examination or specialized testing in clinical care: Asymptomatic screening, leading
can reveal subtle or even marked impairments in to a clinical diagnostic workup if screening phys-
nerve structure and function. With progression to ical examination results are abnormal, or alterna-
foot muscle weakness (motor weakness repre- tively through complaints of neuropathic
sents another large fiber dysfunction), or clinical symptoms on a clinical encounter, leading simi-
autonomic manifestations like dry feet from lim- larly to a clinical workup involving history, phys-
ited sweat production (sudomotor dysfunction), ical examination, simple laboratory tests, and

Table 1  Conventional diabetic distal symmetric polyneuropathy (diabetic DSP) staging system with notes on diagnos-
tic considerations
Stage Description and considerations
Stage 0 No diabetic DSP
Absence of neuropathic symptoms, signs, and normal NCS.
In clinical practice, such patients would not be evaluated by NCS, and thus Stage 0 would rarely be
confirmed. This is more relevant to clinical research.
May be associated with dysfunction in objective tests other than NCS:
 Small fiber morphology tests: Examples include intra-epidermal nerve fiber density or other
parameters, subtle abnormalities in corneal nerve fiber density
 Small fiber function tests: heart rate variability, sudomotor dysfunction, abnormal vascular reactivity
to heating or other stimuli
 Large Fiber function tests: subtle abnormalities in monofilament sensitivity
 Large Function morphology tests: Subtle abnormalities in sural nerve biopsy
 Measureable subtle dysfunction in these tests could serve as predictive index tests during Stage 0 for
future onset of diabetic DSP. Proof-of-concept exists for the 8-point distal monofilament examination
and for corneal nerve fiber density [8] (CNFL in press)
Stage 1 Asymptomatic diabetic DSP
Stage 1a Absence of neuropathic symptoms, signs, but with abnormal NCS
In clinical practice, such patients would not be evaluated by NCS and thus this stage would rarely be
identified. This is more relevant to clinical research
Stage 1b Abnormality in nerve conduction studies and neuropathic signs, but absence of neuropathic symptoms
In clinical practice, such pateints would be identified by asymptomatic screening as recommended by
guidelines
These subclinical stages 1a and 1b could similarly be associated with abnormalities in other large fiber
morphology and function measures, or in small fiber measures as outlined for Stage 0
Clinical Diagnosis of Diabetic Peripheral Neuropathy 69

Table 1 (continued)
Stage Description and considerations
Stage 2 Clinical diabetic DSP
Stage 2a Criteria for subclinical neuropathy have been met and there are neuropathic symptoms with or without
neuropathic signs
In clinical practice, such people would be identified based on symptomatic complaints leading to a
history and physical examination
At this stage, neuropathy is sufficient for complications including neuropathic pain and sensory ataxia
(imbalance)
Stage 2b Criteria for subclinical neuropathy have been met and there is unequivocal weakness of ankle
dorsiflexion
Beyond this initial motor manifestation change, later stages (undefined by this classification scheme)
include progressive motor dysfunction and the presence of foot complications such as ulceration,
infection, amputation, and the Charcot deformity
DSP distal symmetric polyneuropathy, NCS nerve conduction studies
Table modified on content in Dyck et al. [9]

consideration in a small subset of people for neu- type 2 diabetes, the existence of pre-diabetes may
rologist referral for specialized testing and lead to polyneuropathy and therefore it may be
evaluation. present at diagnosis even without substantial ele-
vations in glycemic exposure [17].

2 Key Clinical Considerations


FOR Diabetic DSP Diagnosis 2.2 The Impaired Protective
Sensation of Diabetic DSP
2.1 Clinical Risk Factors Represents Only One
for Diabetic DSP Are Typically Component Cause of Foot
Present Complications

Elevated cumulative glycemic exposure is the While diabetic DSP itself is sufficient to cause
fundamental risk factor for diabetic DSP onset pain and imbalance as complications, it repre-
[10–12]. As such, age, diabetes duration, and gly- sents only a component cause, neither necessary
cemic exposure measured by an inability to nor sufficient, to cause ulcer, infection, the
maintain glycated hemoglobin A1c at target lev- Charcot deformity, or amputation. Tissue isch-
els, generally considered to be 7.0% or less, are emia from peripheral vascular disease, and foot
related risk factors [13]. Additionally, cardiovas- deformity or the minor trauma to skin that incites
cular metabolic risk factors including presence of ulceration and infection induced by inadequacies
hypertension, abdominal obesity, hypertriglycer- in footwear and general foot care represent the
idemia, and low high-density lipoprotein, as well other component causes. The implication is that
as taller height, alcohol abuse, and chronic kid- if a clinician is to meaningfully help to prevent
ney disease have been identified, in addition to foot complications, the annual foot evaluation
putative racial and genetic factors that may be must not focus only on the identification of the
clinically identifiable in future [13–16]. However, loss of protective sensation representing diabetic
specific threshold levels of risk factors and their DSP alone. It must include evaluation for arterial
duration have not been proposed. Generally, an patency (pedal pulses and skin changes), and foot
individual with short duration of type 1 diabetes inspection for presence of abnormalities such as
is extremely unlikely to have diabetic DSP unless callouses or deformity that may indicate the pres-
exceedingly high glycemic exposure has occurred ence of repetitive minor trauma (Fig.  1 and
and other risk factors are present. However, in Table 2).
70 B. A. Perkins and V. Bril

Diabetic Neuropathy alone is


Neuropathy sufficient for pain
(Loss of Protective and imbalance
Sensation) (sensory ataxia)

Component Causes
Peripheral for Foot
Vascular Complications
Disease Amputation
(Pain, Ulcer,
(Impaired Tissue Infection, Charcot
Perfusion) deformity)

General Foot
Care
(Deformity and
Minor Trauma to
Skin)

Fig. 1  A simplified view of the core component causes of the presence of peripheral vascular disease or minor
foot complications and amputation. While sufficient for trauma to skin. The implication is that clinical evaluation
pain and sensory ataxia, the presence of loss of protective focused only on diabetic neuropathy, without consider-
sensation from diabetic neuropathy alone is not consid- ation of the other component causes through simple
ered sufficient for advanced foot complications without inspection and palpation for pulses, is insufficient

Table 2  Clinical manifestations according to fiber classification and other key clinical features of diabetic DSP
Fiber classification Large fibers: myelinated Aδ sensory, Aγ motor, Small fibers: thinly-myelinated autonomic
and Aα and Aβ mixed motor and sensory B-fibers, and thinly- or un- myelinated
autonomic and sensory C-fibers
Function Touch, pressure, vibration, proprioception/ Pain sensation, temperature sensation,
balance, somatic motor autonomic
Symptoms Numbness, tingling, unsteadiness Pain, discrimination of hot and cold
Signs Impairments in ankle reflexes, vibration, light Impairments in thermal discrimination,
touch, 10-g monofilament, proprioception, pinprick sensation
sensory ataxia (positive Romberg test) [18]
“Red Flags” that are Asymmetry
inconsistent with Nonlength dependence
diabetic DSP Acute or subacute rather than insidious, chronic onset and progression
Motor predominance [19]
Also consider
Family history of sensory and motor symptoms
Sensory neuropathies causing gait ataxia and proprioceptive dysfunction
Severe dysautonomia [20]
Late stage findings Dry skin (sudomotor neuropathy), callus, fissures, ulcerations, cellulitis
Charcot deformity
Physical examination Inspection for deformity
for other foot  Callus, abrasions, hallux valgus
complication Vascular
component causes  Pallor, cyanosis, capillary refill, pedal pulses (posterior tibial and dorsalis pedis)
Diabetic DSP diabetic distal symmetric polyneuropathy
Adapted from concepts introduced by Pop-Busui et al. [1]
Clinical Diagnosis of Diabetic Peripheral Neuropathy 71

2.3 Impaired Protective Sensation 3 Asymptomatic Clinical


Shows Heterogeneity Presentation: Screening
Between People with Diabetes for Diabetic DSP

Impaired protective sensation may occur asymp- It is estimated that approximately half of people
tomatically (in approximately half of those with with objective evidence by physical examination
objective evidence of diabetic DSP) [21] or symp- or specialized testing have no symptoms of neu-
tomatically. Whether symptomatic or not, the ropathy [1, 21]. Asymptomatic screening for neu-
symptoms and signs may represent small or large ropathy has therefore generally been justified
fiber sensory modalities, as well as autonomic and based on “disease principles” criteria for screen-
motor modalities. Severity can be determined by ing [23]. For example, the epidemiology of dia-
the extent of sensory modality impairment, and by betic DSP is generally understood, its natural
the presence and extent of motor weakness in the history includes a latent phase, and the target
muscles of the feet. Furthermore, while autonomic population for screening—including type 2 diabe-
dysfunction can be observed at early stages by tes at diagnosis and type 1 diabetes with 5 or more
specialized testing, clinical autonomic manifesta- years of diabetes duration—is well defined [1].
tions generally occur at more advanced stages. Screening is also supported by “test principles.”
Finally, many may present with mixed findings For example, screening test performance charac-
representing both small- and large-fiber modalities teristics and their interpretation and thresholds are
[1, 11]. The implication of this heterogeneity of generally well understood [23]. This justification
clinical presentation is that the evaluating clinician exists even if “system principles” are not uni-
must consider these varied attributes, that theoreti- formly met. While a diabetes multiprofessional
cally the selection of individual screening tests care infrastructure exists, screening is acceptable
may not be sufficient, and that scales that consider to people with diabetes and carries little harm,
multiple attributes may have advantages over other some aspects are not yet well understood. These
investigative tests. include how valid specific tests are when imple-
In summary, evaluation of the feet in people mented in different settings, the economic evalua-
with diabetes represents consideration of risk tion of screening, the lack of neuropathy-­specific,
factors for diabetic DSP, evaluation of a com- and the implications of misclassifying neuropathy
posite of the varied components of diabetic as present when it is not (false positive) or falsely
DSP, as well as a composite of the features of negative. Despite these limitations, the diabetes
neuropathy, peripheral vascular disease, and community has consistently supported screening
foot deformity and foot care. The diagnosis of for diabetic DSP [1, 24, 25].
diabetic DSP itself requires a process of clinical In practice, the annual screening examination
assessment, either initiated by asymptomatic recommended by diabetes organizations can be
screening or in response to a symptomatic com- accomplished by way of very simple tests for
plaint, that considers the results of various clini- loss of protective sensation. Examples include
cal history and examination tests. As in any testing pressure sensation with a monofilament,
process of diagnostic medicine, each symptom testing vibration sensation with a tuning fork, or
or sign incrementally revises the clinician’s esti- frankly an even simpler “Touch the Toes”
mates of disease probability and reduces clini- approach adopted by Diabetes UK in which the
cal undertainty [22]. feet are simply exposed, inspected, and touched
72 B. A. Perkins and V. Bril

with the fingertip [24, 26, 27]. Some organiza- tional multi-site testing, in the test variation the
tions, like the American Diabetes Association monofilament is applied four times to each great
through its neuropathy position statement, rec- toe in a random, arrhythmic manner to generate
ommend a more extensive evaluation. This rec- a quantitative score. For each stimulus, a score
ommendation calls for an annual careful of 0 is assigned if it is not perceived, 0.5 if it is
neuropathic history, along with a small fiber substantially less than that perceived on the
function test (such as temperature or pinprick forehead or sternum, and 1 if it is perceived nor-
sensation), a large fiber test (vibration sensation mally, resulting in a total score from 0 (com-
using a 128-Hz tuning fork), as well as a simpli- pletely insensate) to 8 (completely sensate).
fied version of the 10 g monofilament test to Unlike the binary score, this allowed determina-
assess for feet at risk of ulceration or non-trau- tion of two specific thresholds: The concurrent
matic amputation [1]. validity threshold for identifying the presence of
While we discuss physical examination neuropathy in cross-sectional study (this was
maneuvers below under Neuropathic Signs, we determined to be a score of 0–3 out of 8), as well
wish to discuss a particular aspect of screening as a predictive validity threshold not associated
for impaired protective sensation using common with the current presence of neuropathy but
maneuvers. Most maneuvers have been justified instead with future onset in a 4-year longitudi-
by their face validity: They make sense, they are nal cohort structure (this was determined in a
available in clinical practice, they are well rec- longitudinal cohort to be a score of 3.5–5.5). A
ognized by diverse clinicians, and represent a score of 6–8 is interpreted as ruling out current
key attribute of diabetic DSP. Several common neuropathy and extremely low 4-year risk of
examples exist that have more extensive research onset [8, 24]. In summary, while frank loss of
evidence in that they not only have demon- sensation to the 10g monofilament helps to pre-
strated face validity and reasonable test repro- dict future ulcer and amputation risk among
ducibility, but also intrinsic diagnostic accuracy those with neuropathy, a quantitative variation
for concurrent validity [28, 29]. Concurrent of this examination seeking more subtle changes
validity represents the ability of a test to reason- in sensation that would otherwise be called
ably classify, in a cross-sectional study design, “normal” can be used as a screening tool, such
who in the study sample has neuropathy defined that asymptomatic individuals (Stage 0 and 1a)
by a reference standard test (including nerve can be reasonably stratified into those at low and
conduction studies) and who does not. Several high future risk. This, after all, is the intention
examples of tests with this level of evidence of asymptomatic screening for disease: to iden-
exist [28, 29]. However, we highlight a particu- tify at the earliest stages when interventions,
lar quantitative variation of the 10 g-monofila- such as improvement in glycemic control, are
ment examination that has even further evidence more likely to be effective. The identification of
as a screening test for the prediction of future other tests, including objective measures, that
onset of diabetic DSP. This scoring system has could have sensitive thresholds to identify indi-
been created in order to measure more subtle viduals at future risk of neuropathy onset repre-
degrees of abnormality in protective sensation sents an urgent clinical research need [30].
[8, 24]. First, it is applied only to a distal, non- While the more sensitive, quantitative variation
callousing site (proximal to the nail bed as of the monofilament examination provides an
shown in Fig. 2) in order to identify earlier, dis- approach to screening for protective sensation
tal length-dependent sensory impairment as that has been evaluated in meta-analysis [28],
compared to the more traditional multi-site test- we readily acknowledge that the predictive
ing that includes more proximal sites on the validity findings arise form a single-­center lon-
soles of the feet. Second, rather than simply gitudinal cohort and that it is not known with
grading a 10g monofilament test as a binary certainty if there are advantages over other sim-
“normal” or “abnormal” result as in the tradi- ple [27] or complex [1] screening approaches.
Clinical Diagnosis of Diabetic Peripheral Neuropathy 73

Fig. 2  Quantitative variation of the 10 g monofilament the skin, bend the filament for a full second, then lift from
test suitable for diabetic dsp screening for the presence of the skin. (5) Perform this stimulus four times per foot in
neuropathy (impaired protective sensation) or its future an arrhythmic manner so the person does not anticipate
risk. The 10g monofilament is also known as the 10 g when the stimulus is to be applied. (6) For each of the 8
Semmes-Weinstein monofilament. Adapted from Bril stimuli, assign a score of 0 if it is not perceived, 0.5 if it is
et al. [24] The testing procedure: (1) Show the 10 g mono- substantially less than that perceived on the forehead or
filament to the person with diabetes. (2) Touch it first to sternum, and 1 if it is perceived the same. A score of 3 out
the person’s forehead or sternum so that the sensation is of 8 correct responses means that the presence of neuropa-
understood. (3) Instruct the person to say “less” or “same” thy is likely. A score of 3.5–5 means that the risk of new-­
every time the monofilament stimulus is perceived. (4) onset neuropathy in the next 4 years is high. A score of 5.5
With the person’s eyes closed, apply the monofilament to or greater indicates that there is a low risk of neuropathy
the dorsum of the great toe proximal to the nail bed as onset in the next 4 years
shown in the illustration. Use a smooth motion to touch

4 Neuropathic Symptoms clinically informative as there is a subtlety to the


lived experience of polyneuropathy [31, 32].
The common symptoms of diabetic DSP are the Some recommend classification by small and
large and small fiber sensory manifestations. large fiber sensory modalities [1], and we describe
Historically classified as “positive” symptoms, clinical features according to this classification
indicating nerve hyper-function creating a sensa- throughout this chapter (summarized in Table 2).
tion normally absent, or “negative” symptoms, Regardless of classification, common symptoms
that represent hypo-function creating loss of a are pain, tingling, numbness, unsteadiness, and a
sensation normally present, this distinction is not feeling of weakness. Symptoms typically have a
74 B. A. Perkins and V. Bril

slow, insidious progression, beginning bilaterally feels exaggerated, while allodynia refers to a nor-
at the tips of the toes moving proximally over mal stimulus that is perceived as painful such as
months or years to involve the lower thighs at severe pain induced by rubbing the toes along the
which point the hands may become involved. bedsheets. While uncommon in diabetic DSP
This “stocking and glove” distribution of sensory without first experiencing sensory symptoms,
symptoms can even progress to involve the mid- symptoms of autonomic dysfunction such as dry-
line of the abdomen, and symptom descriptions ness of skin from sudomotor neuropathy, or the
are variable between people. Unequivocally, symptoms of gastroparesis, enteropathy, or clini-
symptoms present in the hands that are not cal cardiac autonomic neuropathy with postural
accompanied by clinical features first in the lower lightheadedness and syncope may occur. Finally
limbs at or below the knee are not consistent with the symptoms of foot complications like infec-
a diagnosis of diabetic DSP.  Among the small tion, ulceration and Charcot deformity may be
fiber symptoms, common pain descriptors are present symptoms in advanced neuropathy.
“burning,” “stinging,” “shock-like,” lancinating
pain [33]. Less common descriptors are a
“squeezing,” pressure sensation. Pain can be 5 Neuropathic Signs
present during the daytime or nighttime, and
often have paroxysmal patterns. Small fiber dys- A directed neurological examination for diabetic
function also includes impaired temperature dis- sensorimotor polyneuropathy (DSP) requires
crimination, often described as a loss of ability to examination of the lower limbs starting distally at
identify hot or cold water with one’s feet in the the toes [33]. As with symptoms and depending
bathtub. Large fiber sensory symptoms include on stage and severity, the signs may be a reflexion
tingling, “pins and needles,” or numbness. of small fiber dysfunction, large fiber sensory
Numbness can represent a lack of sensation, and dysfunction, large fiber motor weakness, pres-
is often used variably by people with diabetes to ence of clinical autonomic abnormalities such as
describe lack of thermal sensitivity, light touch, sudomotor neuropathy, and the manifestations of
lack of pain sensation from minor traumas that foot complications (Table 2). Appearance of the
would normally be present, such as the finding of skin and feet can reveal dryness and color
a stone in one’s shoe or a cut to the skin without changes, loss of hair, clawing and deformity of
feeling pain or discomfort. People frequently the toes, and ulcerations at late stages. Clinical
describe a relative feeling of unsteadiness, which sensory function is assessed for small (thermal
can be a manifestation of pain, tingling, numb- sensitivity, pin prick), large (vibration, proprio-
ness, or instead from a sensory ataxia in which ception), and mixed (light touch) fiber modali-
either foot proprioception is affected or the stabi- ties. These sensory tests are performed by first
lizing effect of feeling the ground. Similarly, a applying the stimuli to the sternum or forehead so
nonspecific feeling of weakness is a very com- that the person can appreciate the normal sensa-
mon descriptor, likely as a consequence of abnor- tion, and then distally at the first toe, or adjacent
mal sensory symptoms, as frequently this is toe, in case of amputation. To determine severity,
described even when motor testing is completely the stimuli are then moved proximally to provide
normal. Motor symptoms resulting from muscle a level at which the sensory function becomes
weakness can occur in more advanced diabetic normal. For vibration, a 128  Hz tuning fork is
DSP with impairment of foot dorsiflexion and sufficient and must be applied on bony surfaces.
plantar flexion that may be described as an inabil- Alternatively, small pocket-sized battery-­
ity to perform activities that require standing on operated electronic devices that standardize
the forefoot or on extended toes or heal-walking. vibration can be used [34]. For proprioception,
Hyperalgesia and allodynia as manifestations of the distal phalanx is held on the lateral surfaces
abnormal sensory processing are less common. and moved upwards and downwards in small
Hyperalgesia refers to a painful stimulus that movements. For pinprick, a disposable sterile
Clinical Diagnosis of Diabetic Peripheral Neuropathy 75

sharp metal tip can be used [35]. For thermal sen- well as by the performance of objective confir-
sitivity, cold sensation can be assessed with a matory tests if diagnostic uncertainty remains
steel tuning fork cooled under running cold (chapter “Diagnostic Techniques for Diabetic
water, or alternatively, with a simple pen-like Peripheral Neuropathy”).
device that has a cool metallic end and a warmer Though research into implementation is lim-
plastic end for comparison [36]. For light touch, ited, we make brief mention of certain point-of-­
a cotton wisp may be used. The 10 g monofila- care devices, defined as simple, rapid, lab-quality
ment is a nylon thread affixed to a handle that diagnostic devices for use by clinicians at the
bends at a standardized pressure of 10 g, moment of clinical care for diagnosis. Some are
­representing a combination of touch and pressure an extension of the physical examination (battery-­
sensation. Other physical examination tests such operated vibrating device in place of a tuning
as 2-point discrimination, or the tactile circum- fork, temperature discrimination devices, or the
ferential discriminator to evaluate a person’s abil- “Neuropad” paper to identify sweat function)
ity to differentiate stimuli of different diameters, [34–36, 38, 39], and, like sensory physical exam-
are not commonly used for assessment of dia- ination, many are heavily dependent on subjec-
betic DSP. tive patient responses. Objective sudomotor
Motor function is evaluated by muscle bulk function assessment by way of electrochemical
and power of the small foot muscles, and specific skin conductance devices have shown conflicting
strength on foot dorsiflexion and plantarflexion evidence in the literature [40–43], but may be
are generally tested. Strength in other lower limb amenable to implementation into practice for
groups should also be assessed (knee and hip screening [44]. Simplified point-of-care nerve
movements). The deep tendon reflexes need to be conduction devices for sural nerve amplitude and
performed with the person fully relaxed and knee conduction velocity are reproducible and valid,
and ankle reflexes should be assessed. Gait and and in future may have impact on clinical
ability to perform a tandem gait, Romberg test for decision-­making as well as implementation into
sensory ataxia, and also ability to walk on heels practice [45–49].
and toes would complete the lower limb exami-
nation. Upper limbs should be examined depend-
ing on lower limb findings [33]. 6 Composite Symptom Scales,
From a diagnostic perspective, the principal Sign Scales, and Combined
purpose of the physical examination is to deter- Scales
mine, through the process of clinical assessment,
if the various signs are in keeping with the clini- We have introduced thus for in this chapter the
cal pattern of diabetic DSP through a process, essential components of clinical evaluation,
with each finding, of incrementally revising clini- mostly representing symptoms and signs that
cal estimates of disease probability. For example, represent effective clinical practice but that have
as discussed later, signs such as motor predomi- not individually been evaluated in investigative
nance over abnormal sensory tests, asymmetry, test research for their diagnostic performance.
and lack of length-dependence are findings that However, we then introduced some specific
substantially decrease the probability of diabetic screening maneuvers that have concurrent valid-
DSP. This is of particular importance because it ity for identifying the presence of DSP such as
is known that individual physical examination the Ipswich Touch Test adopted by Diabetes UK
signs are known to have very poor inter- and [27] and versions of pain, vibration, light touch
intra-rater reproducibility, even in the hands of sensation [28, 29], as well as the example of a
expert neurologists [37]. This can be partially variation of the 10-g monofilament examination
overcome by the clinician having a broad, sys- that may permit prediction of future onset of DSP
tematic physical examination including multiple [8]. Investigative test research is broad, though,
physical examination maneuvers as described, as as these tests can include single symptoms, signs,
76 B. A. Perkins and V. Bril

laboratory parameters, as well as composite scor- unscored, including generalized weakness and
ing systems [50]. As individual symptoms or cramping, that can help the rater understand other
signs are likely not sensitive or specific enough to features that may not relate directly or specifi-
serve as a biomarker response, for example, to a cally to DSP and foot complications risk factors.
therapeutic intervention in a clinical trial, one The second component is a five-part lower
strategy is to implement a composite score, such extremity physical examination made up of
as a self-reported symptom questionnaire or the examination for abnormal appearance including
clinical scales that make up a focus of this presence of callus or deformity, examination for
Chapter [51]. While these have been developed ulceration, ankle reflexes, and simple 3-level
primarily as a way to summarize the proportions scores using vibration sensation and the mono-
and severity of diabetic DSP in trial or cohort filament (present, reduced, absent).
study populations, these composite scales can
serve four key purposes to the clinician [22, 50].
First, they incorporate (and remind the clinician 7.1 Scale Face Validity
of the) multiple aspects of the manifestations of
diabetic DSP, rather than focusing on one in iso- Both the MNSI Questionnaire and MNSI
lation. Second, they can provide a diagnostic Examination scales incorporate attributes that relate
threshold for concurrent diagnosis, and, though specifically to polyneuropathy, however, they test
they have not yet been evaluated in this way, for the presence of other attributes including the pres-
prediction of future onset of diabetic DSP or its ence of foot deformities. Whether the person has
related foot complications. Third, the quantitative polyneuropathy or not, the presence of foot abnor-
scores allow a measure of diabetic DSP severity malities such as callus, fissures, ulceration likely
that is typically not conferred by the result of a places them at high risk of foot complications.
single symptom or sign. Finally, many identify Though less specific to diabetic DSP, these features
pain and its severity and can therefore be used to increase its clinical relevance as an outcome mea-
guide therapy for painful diabetic DSP. sure. For polyneuropathy-­ specific research, the
manifestations especially on foot examination may
not be sensitive enough to identify those at earlier
7 The Michigan Neuropathy stages of polyneuropathy alone, given that the sen-
Screening Instrument (MNSI) sory tests are graded in three simple categories.

First published in 1994 [52], the Michigan


Neuropathy Screening Instrument (MNSI) has 7.2 Test Quality and Reliability
had major impact on the identification of diabetic
DSP, as well as providing a measure of its sever- The MNSI itself has not been tested formally
ity, and, potentially also an ability to track pro- using standard methods and metrics for inter- and
gression and regression in many observational intra-rater reproducibility. Studies on variations of
studies and clinical trials [21, 53–56]. The MNSI the MNSI show suitably high reliability [57, 58].
is, in fact, made up of two separate assessments.
First, a questionnaire (symptom scale) compo-
nent, has major value for screening as it is a self-­ 7.3 Diagnostic Accuracy:
administered questionnaire made up of 15 items. Concurrent
These include questions such as having received
a diagnosis by a physician in the past, or a history The original publication, representing analysis of a
of amputation or ulceration, dry skin, but also small cross-sectional group of type 1 and type 2 par-
about neuropathic symptoms such as numbness, ticipants who had diabetic DSP confirmed by nerve
pain, temperature sensation, and allodynia. conduction studies, undertook a simple tabulation
Furthermore, there are two questions that are suggesting that an MNSI Questionnaire score of 7
Clinical Diagnosis of Diabetic Peripheral Neuropathy 77

or greater, and an MNSI Examination score exceed- with improved outcomes relative to current stan-
ing 2, are associated with extremely high predictive dards of screening and diagnosis. However, it has
values positive, good sensitivity and extremely high been frequently operationalized as a neuropathy
specificity [52]. However, a diagnostic study design outcome in large-scale clinical trials including
conducted in a type 1 diabetes observational cohort those evaluating cardiovascular and microvascu-
that also operationalizing nerve conduction studies lar outcomes in type 1 and type 2 diabetes [21,
to define ­diabetic DSP cases, suggested that these 53–56]. As many of these studies required a mea-
particular thresholds were associated with substan- sure of more advanced diabetic foot disease, the
tially lower diagnostic performance, and suggested MNSI has provided a successful proof of concept
alternate thresholds [51]. First, a threshold score of for applying neuropathy scales in trials.
4 or greater on the MNSI Questionnaire, such a test
result would carry 40% sensitivity and 92% speci-
ficity. Second, an MNSI Examination result exceed- 8 Toronto Clinical Neuropathy
ing a score of 2 carried 61% sensitivity and 79% Score (TCNS)
specificity. However, on inspection of the receiver
operating characteristic curves from this publica- The Toronto Clinical Neuropathy Score (TCNS),
tion, we observe that an MNSI Questionnaire score first validated and reported in 2002, was devel-
(such as a score of 1 or greater) could reasonably be oped specifically for use in a study of simple
chosen to maximize sensitivity to nearly 60%. screening measures for DSP that could be used in
the family physician’s office or the diabetes clinic
[29, 59]. Spectrum bias is a very common design
7.4 Diagnostic Accuracy: flaw (a source of selection bias) in investigative
Predictive test research. It represents inappropriate recruit-
ment of many participants for whom there is no
While many longitudinal designs have been con- diagnostic uncertainty, and recruitment of partici-
ducted, and even some cohort designs (in which pants with limited variation in disease characteris-
those with neuropathy at baseline have been tic [22]. The original purpose of the TCNS was to
excluded), we were unable to find in the literature create a quantitative score that ranged across all
analyses that explored whether a certain test diabetic DSP Stages (Table  1) that would allow
score could predict future onset of diabetic for stratified accrual across the full spectrum of
DSP. While this does not make inherent sense for neuropathy to appropriately determine the diag-
the MNSI examination scores, which measure nostic performance of simple screening measures
manifestations of established diabetic DSP (such [29]. The scale comprises six symptoms (present/
as presence of ulcer or complete reduction or absent), knee and ankle reflexes (normal, reduced,
abnormality—rather than subtle impairments— or absent scored bilaterally) and sensory tests
of vibration or monofilament sensitivity), such a (normal/abnormal) as shown in Table 3, Panel A
design could be applied to the MNSI and ranges from a perfectly normal score of 0
Questionnaire that has components that are suited without clinical evidence of DSP to a maximum
to earlier, pre-diagnostic stages of diabetic DSP. abnormal score of 19 points. The symptoms are:
foot pain, numbness, tingling, weakness, unsteadi-
ness, weakness, and presence of upper limb
7.5 Effect on Treatment Decisions, symptoms. The sensory signs include sensory
Impact on Patient Outcomes, tests of pinprick, temperature, light touch, vibra-
Economic Analysis tion, and proprioception. The deep tendon reflexes
include the knee and ankle reflexes. Out of a total
The results of MNSI testing have not been studied score of 19, the grades are defined as follows: 0–5
on their effect on the treatment decisions made by = no neuropathy; 6–8 = mild neuropathy; 9–11 =
clinicians, or whether MNSI testing is associated moderate neuropathy; ≥ 12 = severe neuropathy.
78 B. A. Perkins and V. Bril

Table 3  The components of the original Toronto Clinical 8.2 Test Quality and Reliability
Neuropathy Score (TCNS) and the modified TCNS
(mTCNS)
Formal evaluation of reliability reported internal
Sensory test Reflex
Symptom scores scores scores
consistency of attributes, and very good to excel-
A. Scoring of the Toronto clinical neuropathy score lent inter- and intra-rater reproducibility [60].
(TCNS) While in opposition to the findings of poor repro-
Foot pain Pinprick Knee ducibility of individual physical examination
Numbness Temperature reflexes [37], reproducibility is likely greater with investi-
Tingling Light touch Ankle
Weakness Vibration reflexes
gative tests that make up a composite of multiple
Unsteadiness Position sense Reflexes individual tests.
Upper limb Sensory test graded as
symptoms scores graded as  0 =
Symptom scores  0 = normal normal
graded as  1 = abnormal  1 =
8.3 Diagnostic Accuracy:
 0 = absent reduced Concurrent
 1 = present  2 =
absent Diagnostic accuracy has been evaluated exten-
Bilaterally
sively, including relationship with nerve conduc-
Maximum TCNS Score 19 points
tion studies, diabetic DSP classified by signs,
B. Adaptations in scoring to create the modified TCNS
(mTCNS) symptoms, and nerve conduction studies, and
Symptom scores Sensory test Not scored even relative to the results of fiber density on
graded as scores graded as sural nerve biopsies [59, 60]. Furthermore, its
 0 = absent  0 = absent (0) validity has been tested in non-diabetic forms of
 1 = present  1 = present at
without interfering level of the polyneuropathy [61]. In a type 1 diabetes cohort,
with sense of toes TCNS had receiver operating characteristic curve
well-being or  2 = present at area under the curve of 0.86 for identification of
activities level of the Stage 2 diabetic DSP, with an optimal threshold
 2 = present and ankles
interfering with  3 = present at value of 6 or greater identifying DSP with sensi-
well-being but not a level tivity and specificity each approximating 80%
activities proximal to [51]. Additionally, very good overall accuracy
 3 = present and the ankles was achieved even for the identification of sub-
interfering with
well-being and clinical Stage 1a neuropathy, with an area under
activities the curve of 0.74 [62]. However, its diagnostic
Maximum mTCNS score 33 points performance in type 2 diabetes, along with objec-
Adapted from Bril et al. [59, 60] tive tests, was lower [52].

8.1 Scale Face Validity 8.4 Diagnostic Accuracy:


Predictive
The TCNS has attributes that relate specifically
to manifestations of small and large fiber dys- A pilot longitudinal study that included baseline
function of polyneuropathy, excluding other TCNS indicated subtle baseline differences in
components such as foot deformity that raise risk scores between those with future incident diabetic
for foot complications. The broad scoring range DSP and those who remained neuropathy-­free,
that includes six points for symptoms and 13 for but it did not appear to have diagnostic predictive
signs may permit greater sensitivity for earlier validity [63]. Though not conclusive, this implies
stages, though individual test components are that the main role of the TCNS, like other scales,
assigned binary or three-level ordinal scores is optimized for identification rather than on pre-
(Table 3). diction of future neuropathy incidence.
Clinical Diagnosis of Diabetic Peripheral Neuropathy 79

8.5 Effect on Treatment Decisions, fers a measure of earlier stages of diabetic DSP,
Impact on Patient Outcomes, and expands the scoring of symptoms and sensory
Economic Analysis signs to include test components that are assigned
four-level ordinal scores each (Table 3, Panel B).
Similar to the MNSI, the TCNS has not been
studied for effect on the treatment decisions made
by clinicians or screening and diagnosis out- 9.2 Test Quality, Reliability,
comes, but it has frequently been operationalized and Accuracy
as a neuropathy outcome in clinical trials and
cohort studies [44, 64–68]. The trials, however, The very good to excellent inter- and intra-rater
are studies of neuropathy therapies as opposed to reproducibility of the mTCNS was formally eval-
the large-scale cardiovascular outcome trials that uated and reported along with a new evaluation
the MNSI has been used as a neuropathy out- of the original TCNS in a multicenter study [60].
come measure. Furthermore, as hypothesized, scores shared
lower correlation with the TCNS as they measure
different stages of diabetic DSP (Pearson coeffi-
9 Modified Toronto Clinical cient 0.56), but validity for identification of Stage
Neuropathy Score (mTCNS) 2 neuropathy remains similar [60]. However, a
specific threshold has not been independently
With the intention of creating a scale that is more assessed for the concurrent validity of diabetic
sensitive for earlier stages of neuropathy, the DSP, and also ranges of severity have not been
TCNS has been adapted to the modified TCNS assigned as for the TCNS.
(mTCNS). The modified scale omits the deep
tendon reflex assessment, which was a somewhat
less reproducible part of the TCNS [60], and that 9.3 Effect on Treatment Decisions,
additionally represented later (motor) dysfunc- Impact on Patient Outcomes,
tion on the spectrum of diabetic DSP severity. Economic Analysis
The mTCNS also adds greater quantitative levels
to both symptoms and sensory signs to create a Similar to the MNSI and the TCNS, the mTCNS
larger measurement range across the earlier DSP has not been studied for effect on the treatment
stages, and a more granular scale. The symptoms decisions made by clinicians or screening and
of the TCNS are graded according to patient-­ diagnosis outcomes, but it has frequently been
reported outcome principles as absent (0), pres- operationalized as a neuropathy outcome in clini-
ent but not interfering with activities or sense of cal trials and cohort studies that by design focus
well-being (1), present and interfering with sense on earlier stages of diabetic DSP [69, 70].
of well-being but not with activities (2), and pres- Furthermore, it is being implemented in contem-
ent and interfering with activities (3). The signs porary studies of populations at risk of earlier
of the TCNS are graded as absent (0), present at stages of neuropathy (https://clinicaltrials.gov/
the toes (1), present up to the ankles (2), and pres- ct2/show/NCT04664426).
ent proximal to the ankles (3) in a stocking distri-
bution. The mTCNS score ranges from 0 to 33, as
detailed in Table 3, Panel B. 10 Neuropathy Impairment
Score of Lower Limbs
(NIS-LL)
9.1 Scale Face Validity
The NIS-LL is an extensive composite score of
The mTCNS has modifications that, compared to neurological signs and objective tests in the lower
the TCNS, exclude motor manifestations that con- limbs, excluding symptoms, that was designed
80 B. A. Perkins and V. Bril

primarily for implementation in clinical trials 10.2 Test Quality, Reliability,


[71]. This physical examination score was devel- and Accuracy
oped by investigators at the Mayo Clinic as a
simplification of an even more extensive score, The NIS-LL has not been evaluated specifically
termed the NIS-LL+7 that incorporated the for reproducibility or concurrent validity for a
results of seven objective tests including vibra- diagnosis of diabetic DSP. However, it was evalu-
tion perception testing, heart rate variability with ated in a comparative study of neuropathy associ-
deep breathing as a measure of autonomic dys- ated with impaired glucose tolerance subjects
function, and five nerve conduction study param- and in this setting—as with the other scales eval-
eters. This extensive version is considered to uated—its overall diagnostic accuracy was excel-
have 100% sensitivity for Stage 1a diabetic DSP lent [73].
[72]. The NIS-LL clinical scale version (exclud-
ing the seven additional objective tests) incorpo-
rates extensive motor, sensory, and reflex physical 10.3 Effect on Treatment Decisions,
examination maneuvers with granular scoring Impact on Patient Outcomes,
systems. For example, grading of muscle power Economic Analysis
involves eight discrete levels ranging from 0 to 4,
including quarter points. The overall scale is Similar to the previous composite scores, the
graded from a perfectly normal score of 0 to a NIS-LL has not been studied for effect on the
maximum of 88 points [71]. treatment decisions made by clinicians or
screening and diagnosis outcomes as it primar-
ily represents a research tool. However, it has
10.1 Scale Face Validity been extensively applied as a neuropathy out-
come in clinical trials and cohort studies that by
The NIS-LL assigns 64 of the 88 points to muscle design focus on later stages of diabetic DSP
strength attributes that are frequently absent [74–77].
when first clinically identified by clinicians, and
is therefore at face value a measure of later stage
(generally Stage 1b). The potential score for sen-
sory testing is 16/88 points with a maximal loss 11 Utah Early Neuropathy Scale
of sensory modalities of touch pressure, pinprick,
vibration (using the 165Hz tuning fork), and joint This physical examination scale was developed
position. The reflexes at knees and ankles are specifically to serve the need for measurement
considered as present, reduced or absent, and scales that could detect early stages of sensory-­
defined according to age limits adding potential predominant polyneuropathies such as DSP at
confusion to non-expert examiners. Both in terms Stage 1b [78]. The scale includes limited scores
of the later stage diabetic DSP identification and for motor function (great toe extension and
the expertise required for examination, the ankle reflexes), and greater proportional contri-
NIS-LL is not seen as a tool that can be translated bution to sensation for pin and vibration as well
to clinical diagnosis, but rather as a specialize as allodynia. The scale varies from 0 to 42 with
tool for interval cohort studies and clinical trials. 24/42 points for pin sensation, 8/42 for vibra-
Given its wide spectrum of motor scores, at face tion, and 2/42 for allodynia. To demonstrate an
value it might represent a sensitive measure for example of a score for earlier sensory manifes-
neuropathic change in clinical trials examining tations and how it is operationalized, we include
the effect of interventions on more advanced neu- the scoring for the Utah Early Neuropathy Scale
ropathy stages. in Fig. 3.
Clinical Diagnosis of Diabetic Peripheral Neuropathy 81

Utah Early Neuropathy Scale

Motor Examination Right Left

0 normal

2 weak
Great Toe Extension
Total Both Sides (out of 4): Segments for pin sensation reporting

Right Leg Left Leg


Pin Sensation: Right Left
0 normal 6 6

1 for each segment with


reduced sensation 5 5
2 for each segment with
absent sensation 4 4
3 3
Total out of both sides: (out of 24):
2 2
1 1

Allodynia/Hyperesthesia Right Left


0 normal
1 if present In toes or foot

Total for both sides (out of 2):

Large Fieber Sensation Right Left Deep Tendon Reflex Right Left
0 normal 0 normal
1 diminished 1 diminished
2 absent 2 absent
Great toe vibration Ankle
Time s s Total both sides (out of 4):
Great toe joint position

Total both sides (out of 8): Total Score (out of 42)

Fig. 3  Performing the Utah Early Neuropathy Scale instance, a person who reported absent pin sensation to
(UENS) Examination. The UENS requires a number 2 the mid foot dorsum (four points) and reduced sensation
(13/4 inch) safety pin and a 128 Hz tuning fork. Pin sensa- to the low ankle (1 point) bilaterally would score a total of
tion is tested by first reviewing normal sharp sensation to ten points for this portion of the UENS. Vibration is tested
pin on an unaffected portion of the skin. Once this is by first acquainting the subject with vibration (as opposed
established, touch the dorsal surface of the foot and leg to pressure) sensation, then holding the maximally
with the pin, working centripetally from the great toe in vibrated tuning fork to the dorsum of the great toe at the
1–2  cm increments while asking the subject to respond distal interphalangeal joint. Extinction of vibration in less
when they first feel “any sharpness,” and again more prox- than 10 s is considered “diminished,” while “absent”
imally when the pin feels “as sharp as they would expect.” requires that the patient cannot detect the maximally
Repeat to firmly establish these levels. On each side, two vibrating tuning fork at the toe. The motor examination is
points are scored for each region in which the patient fails limited to great toe dorsiflexion. Other aspects are as typi-
to feel any sharpness. One additional point is scored for cally performed in neurological examination. Figure cour-
each additional region in which the pin feels less sharp tesy of A.  Gordon Smith, co-creator of the Utah Early
than expected. Only distal sensory loss is scored. So, for Neuropathy Scale. The Scale is further detailed in [78]
82 B. A. Perkins and V. Bril

11.1 Scale Face Validity 12 Neuropathy Symptom Score


(NSS)
The scale does not include symptoms, and there-
fore in the context of diabetic DSP would be ame- First described in 1993 in a descriptive study of
nable for use as a screening tool to identify Stage the prevalence of the hospital-based clinic diabe-
1b disease. Furthermore, the relative weight of tes population in the United Kingdom [85], the
sensory signs is high, including small fiber func- Neuropathy Symptom Score was an interviewer-­
tion, such that variation of scores likely represent based questionnaire meant to evaluate the pres-
earlier-stage neuropathy when interventions and ence and severity of specific polyneuropathy
therapies are most likely to be of benefit. manifestations, with a particular emphasis on
painful diabetic DSP.  In the interview, patients
are asked about their experience of pain or dis-
11.2 Test Quality, Reliability, comfort in the legs. Characteristics are evaluated
and Accuracy such that burning, numbness or tingling in the
legs provides two points, and fatigue, cramping
Reproducibility has not been evaluated cross-­ or aching 1 point. Location provides 2, 1, and 0
sectionally within and between examiners, but the points for feet, calves, and elsewhere, respec-
1-year change in score showed very high consis- tively. Nocturnal symptoms provide 2 further
tency and has been reported as an indication of points, 1 if also daytime, 0 if only daytime. An
excellent reproducibility [78]. Additionally, it cor- additional point was assigned if patients are awo-
related with other scoring systems. For Stage 2 ken at night from symptoms. Finally, alleviating
polyneuropathy confirmed by nerve conduction factors are scored: 2, 1, and 0 if alleviated by
studies, in a study sample with a very high propor- walking, standing, or laying down. With a maxi-
tion of polyneuropathy participants it showed very mum of 9, it is implied that symptoms are present
good overall concurrent validity by area under the if a minimum score of 3 is achieved. Severity was
receiver operating characteristics curce [78]. This graded as mild (score of 3–4), moderate (score of
level of accuracy appeared to be similar to other 5–6), or severe (score of 7–9).
scales [78], and it has been examined in other
causes of polyneuropathy [79, 80]. However, it is
our view that this scale may show advantage in 12.1 Face Validity
study samples that reflect general diabetes practice,
with lower prevalence of advanced polyneuropathy The score, at face value, weighs heavily on pain-
and higher prevalence of early disease [81]. ful diabetic DSP and its consequences, including
night-time symptoms and awakening, and the
alleviating symptoms in the context of such
11.3 Effect on Treatment Decisions, awakening. Consequently, its primary role is in
Impact on Patient Outcomes, the classification of the presence and severity of
Economic Analysis painful diabetic DSP.

Similar to the previous composite scores, the


Utah Early Neuropathy Scale has not been stud- 12.2 Test Quality and Reliability
ied for effect on the treatment decisions made by
clinicians or screening and diagnosis outcomes While not tested formally for reproducibility,
as it primarily represents a research tool. the NSS has served as a historical benchmark
However, it has been extensively applied as a for the development of updated symptom scores
neuropathy outcome in cohort studies and clini- that correlate highly with it, and that them-
cal trials that by design focus on earlier stages of selves have excellent reproducibility in small
diabetic DSP [82–84]. studies [86].
Clinical Diagnosis of Diabetic Peripheral Neuropathy 83

12.3 Diagnostic Accuracy: 13.3 Diagnostic Accuracy:


Concurrent Concurrent

During the development of the NSS, studies of its During the development of the revised NDS,
intrinsic validity were not conducted. However, it studies of its intrinsic validity were not con-
has been evaluated by independent groups pri- ducted. However, it has been partially evaluated
marily in combination with a physical examina- by independent groups [87, 88]. While there are
tion scale termed the Neuropathy Disability Scale no cross-sectional studies evaluating the optimal
that is discussed below [87, 88]. threshold for the identification of diabetic DSP, a
score of 6 or more has traditionally been used in
studies in which it is the reference standard [89,
13 The Revised Neuropathy 90].
Disability Score (NDS)

Developed as a simplification of the NIS-LL and 13.4 Diagnostic Accuracy:


also first described in 1993 along with the Predictive
Neuropathy Symptom Score in a descriptive
study of neuropathy prevalence, the score incor- While many longitudinal designs have been con-
porates small fiber sensory attribute, and large ducted, we were unable to find in the literature
fiber sensory and motor attributes. It involves analyses that explored whether a certain test
examination of the ankle reflex, vibration, pin-­ score in those without DSP could predict future
prick and temperature (cold tuning fork) sensa- onset of diabetic DSP.  However, among those
tion at the great toe. The sensory modalities are followed in a diabetic foot clinic (and likely had
scored as either present = 0 or reduced/absent = 1 presence of diabetic DSP), the revised NDS was
for each side, and reflexes as normal = 0, present generally associated with short-term risk of inci-
with reinforcement = 1 or absent = 2 per side. dent ulceration, though in this study population
With a maximum abnormal score of 10, classifi- the best prediction was associated with abnor-
cation of severity is graded as mild (score of mality in foot pressure sensation measurement
3–5), moderate (score of 6–8), and severe (score [91]. In combination with the NSS, the revised
of 9 or 10). Generally, a score of 6 or more has NDS has been used by several research groups
been used to define presence of signs for identifi- for the identification of higher risk patients for
cation of DSP [89, 90]. diabetic DSP, and as the clinical sign and symp-
tom measures to define presence of diabetic DSP
[92, 93].
13.1 Face Validity

This score includes small fiber sensory attributes 14 Perspectives


that could identify pre-symptomatic or symptom- on the Comparison of Scales
atic early-stage DSP, as well as later large-fiber
stages owing to sampling of vibration sensation Each of the scales chosen for review in this chapter
and reflexes. has been designed with specific research purposes
in mind. At face value they reflect different aspects
of diabetic DSP (from early sensory signs in the
13.2 Test Quality and Reliability Utah Early Neuropathy Scale, to those that include
advanced foot deformity and ulceration like the
There are no published studies examining intra- Michigan Neuropathy Screening Instrument
and inter-rater reproducibility of the revised Examination). As summarized in the above sec-
NDS. tions and in Table 4, they have different levels of
Table 4  Composite symptom scales, sign scales, and combined scales: general attributes and comparisons
84

Acronym Considerations for face validity Considerations for technical performance


Symptom or Weighted toward Reproducibility Diagnostic Accuracy
signs earlier (sensory) (concurrent validity)
Granularity components or later (motor) For NCS-based
Scale name (range of scores) stages reference standard Other considerations
Michigan MNSI 0–15 Symptoms only Later Not formally Low Can be used as a self-
neuropathy questionnaire assessed administered questionnaire
screening [52] for screening in virtual care
instrument
Michigan MNSI 0–10 Signs Later Not formally Good [51] Operationalized in
neuropathy examination [52] assesseda cardiovascular disease
screening intervention trials [21,
instrument 53–56]
Toronto clinical TCNS [59] 0–19 Both Later Very good to Very good for T1D, Its validation has been
neuropathy score excellent [60] good for T2D extensive, including
agreement with NCS, disease
severity, and sural nerve
biopsy nerve fiber density
[59, 60]
Evaluated in multicenter
study [60]
Modified Toronto mTCNS [60] 0–33 Both Earlier Very good to Correlated with NCS, In a study comparing
clinical excellent [60] not formally assessed multiple scales in the setting
neuropathy score of pre-diabetic neuropathy,
this scale had the highest
numerical sensitivity and
specificity compared to
others [73, 94]
Neuropathy NIS-LL [71] 0–88 Signs Later Not formally Very goodb [75] This represents the signs
impairment score assessed component of the NIS-LL+7,
of lower limbs a scale that includes seven
objective small and large
fiber tests [72]
Diagnostic accuracy
evaluated in pre-­diabetic
neuropathy [73]
B. A. Perkins and V. Bril
Utah early UENS [78] 0–42 Signs Earlier Not formally Very goodb [75] The score incorporates
neuropathy score assessedc multiple levels of sensory
deficit in the lower limbs (see
Fig. 2)
Neuropathy NSS [85] 0–9 Symptoms only Both (pain) Not formally Not formally assessed Interview-based
symptom score assessed on its on its own [87, 88] questionnaire
own Focused on painful
neuropathy and its
consequences
Revised NDS [85] 0–10 Signs Earlier Not formally Not formally assessed Associated with short-term
neuropathy assessed on its on its own [87, 88] risk of ulceration [91]
disability score own
revised
Note that for all scales different diagnostic thresholds have been proposed depending on specific outcomes and specific settings. For example, for screening a more sensitive
threshold is preferred, for diagnosis a more specific threshold is preferred. Many of these scales have been incorporated into reference standard definitions for diabetic DSP that
use confirmatory nerve conduction studies. In those settings, generally a score of only 1 or more is required to support the presence of a symptom or a sign [9]
NCS nerve conduction studies, T1D type 1 diabetes, T2D type 2 diabetes
a
Modified versions have been studied [57, 58]
Clinical Diagnosis of Diabetic Peripheral Neuropathy

b
Study sample had very high proportion with diabetic DSP
c
One-year change was evaluated and reproducibility was high, but generally change over time is a different attribute of ability to detect change in nerve function rather than
reproducibility
85
86 B. A. Perkins and V. Bril

research published on the hierarchy of investiga- ropathies. These include focal nerve entrapments
tive test research principles, studies of validity and other non-distal and non-symmetric forms
include different study samples, study sizes, preva- (chapters “The Epidemiology of Diabetic
lence of neuropathy, spectrum of disease, variable Neuropathy” and “Treatment Induced Neuropathy
reference standard definitions. Even with harmo- of Diabetes (TIND)”), rare conditions such as
nization of such procedures, systematic review hereditary neuropathy with propensity to pres-
and meta-analysis of diagnostic tests are complex sure palsies (HNPP), and polyneuropathies with-
as they require Bayesian techniques like clinical out sensory predominance such as common
trial network metanalysis methods [95]. Though forms of chronic inflammatory demyelinating
we present their attributes and critical appraisal in polyneuropathy (CIDP) and its variants.
detail, for these methodological reasons we inten- Furthermore, consideration should be made for
tionally do not present a quantitative side-by-side factors associated with potentially similar poly-
comparison of their diagnostic operating charac- neuropathy to diabetic DSP such as alcohol,
teristics. One research group has undertaken a hypothyroidism, toxins, and drugs. With this in
direct comparison of many of these scales in the mind, a simple laboratory panel should be under-
setting of pre-diabetic neuropathy and, while they taken: comprehensive metabolic panel to deter-
imply that some are more valid than others (the mine current metabolic risk factors such as
modified TCNS), uniformly in that study most glycemic exposure, presence of hypertriglyceri-
scales had unexpectedly high levels of concurrent demia, renal dysfunction; thyroid hormone to
diagnostic accuracy [73]. We challenge clinicians indicate hypothyroidism; complete blood cell
to consider operationalizing these scales in clinical count may indicate macrocytosis associated with
setting to suit specific needs: For example, the alcohol or nutritional deficiencies and infectious
self-­administered Michigan Neuropathy Screening causes; the B12 level itself; erythrocyte sedimen-
Instrument questionnaire, the Neuropathy tation rate potentially associated with inflamma-
Symptom Scale, or the symptom subscales of tory causes; and serum protein
other composite scores like the modified Toronto immune-electrophoresis to suggest malignancy,
Clinical Neuropathy Score, can be applied for vir- monoclonal gammopathy, or gammopathy asso-
tual diabetes care as has been required during the ciated with rare syndromes like Polyneuropathy,
severe acute respiratory syndrome coronavirus 2 Organomegaly, Endocrinopathies, Monoclonal
(SARS-CoV-2) pandemic, while others could be protein, and Skin lesions (including hemangi-
selected for physical examination screening and oma, hyperpigmentation, hypertrichosis or
evaluation in the clinical setting. Raynaud’s phenomenon as part of the “POEMS”
Syndrome). Testing for infectious causes of poly-
neuropathy should be guided by history, as would
15 Differential Diagnosis: consideration of familial forms of DSP.  It is
A Diagnosis of Diabetic DSP important for the clinician to recognize that the
Requires Consideration patient with a consistent chronic, insidious,
of Other Causes sensory-­predominant distal and symmetric clini-
of Polyneuropathy cal history and physical examination for diabetic
DSP typically only requires this focused labora-
A clinical evaluation consistent with the features tory work-up, and that the most common causes
of diabetic DSP requires consideration of other of DSP other than diabetes are B12 deficiency,
etiologies for distal and symmetric polyneuropa- alcohol, and hypothyroidism, and diabetic DSP
thies. First, the presence of atypical features exacerbated by renal failure. Referral to special-
listed in Table 2 should be identified, as they may ist neurology should be strongly considered in
further indicate other causes of peripheral nerve the uncommon case of atypical features (Table 2),
damage other than polyneuropathy, such as focal, or if rare causes are suspected (Table  5) that
multifocal, or nonlength dependent forms of neu- require confirmatory work-up that specialized
Clinical Diagnosis of Diabetic Peripheral Neuropathy 87

Table 5  An non-exhaustive list of diagnoses on the differential of diabetic distal symmetric polyneuropathya
Metabolic and nutritional disorders
 B12 vitamin deficiency (more common)
 Other vitamin deficiencies: pyridoxine, folate, thiamine, vitamin E
 Malabsorption: Bariatric and gastric surgeries, inflammatory bowel disease
 Renal disease (more common)
 Thyroid disease (more common)
 Acromegaly
Inflammatory
 Chronic inflammatory demyelinating polyneuropathy (CIDP) and its variants (may be more common)
Systemic disorders
 Peripheral arterial disease
 Monoclonal gammopathy/paraproteinemia (more common)
 Amyloidosis
 POEMSb
 Sarcoidosis
 Vasculitisa
 Critical illness polyneuropathy
Infectiousa
 HIV
 Hepatitis B
 Lyme
 Syphilis
 Leprosy
Drugs
 Neurologic and mood: phenytoin, amitriptyline, lithium
 Antimicrobials: nitrofurantoin, metronidazole, chloramphenicol, tuberculosis therapies, chloroquine,
hydroxychloroquine
 Cardiovascular: amiodarone, flecainide, hydralazine, nitrous oxide
 Rheumatic: colchicine, gold, leflunomide, methotrexate
 Immunologic: tacrolimus, interferon-α, ipilimumab, nivolumab, pembrolizumab, bortezomib
 Antineoplastic: chemotherapeutic agents, paclitaxel and other taxanes, vinca alkaloids, platinum analogues,
doxorubicin, etoposide, ifosfamide, misonidazole, antinucleosides
Toxins
 Alcoholism (more common, frequently painful neuropathy)
 Heavy metal: lead, arsenic, inorganic mercury, zinc, thallium, gold
 Herbicides: dichlorophenoxyacetic acid, agent orange
 Organophosphate pesticides: parathion, dioxin
 Industrial: acrylamide, polychlorinated biphenyl, vinyl chloride
 Solvents: hexacarbons (glue sniffing), dry cleaning solvents, carbon disulfide, perchloroethylene,
trichloroethylene, triorthocresyl phosphate, ethylene oxide, styrene, toluene, methyl n-butyl ketone, mixed
solvents, and others
Hereditary
 Hereditary neuropathy with propensity to pressure palsies (HNPP)a
 Multifocal acquired demyelinating sensory and motor (MADSAM) neuropathy
 Other hereditary motor, sensory and autonomic neuropathies
a
Some causes listed here are unlikely to present symmetrically. Differential diagnosis lists in the literature frequently
include causes of non-distal and non-symmetrical neuropathies, such as focal entrapment syndromes and other etiolo-
gies discussed in chapters “The Epidemiology of Diabetic Neuropathy” and “Diagnostic Techniques for Diabetic
Peripheral Neuropathy”. We have made an attempt to minimize these conditions on this list, but those indicated with an
asterisk are potential exceptions
b
Polyneuropathy, organomegaly, endocrinopathy, monoclonal protein, skin abnormalities
88 B. A. Perkins and V. Bril

testing might clarify (chapter “Diagnostic lower-extremity amputation in the young and
middle-­aged adult U.S. population. Diabetes Care.
Techniques for Diabetic Peripheral Neuropathy”). 2019;42(1):50–4. (In eng). https://doi.org/10.2337/
dc18-­1380.
7. Hussain MA, Al-Omran M, Salata K, et al. Population-­
16 Concluding Overview based secular trends in lower-extremity amputation
for diabetes and peripheral artery disease. CMAJ.
2019;191(35):955–61. https://doi.org/10.1503/
The clinical diagnosis of diabetic DSP, initiated cmaj.190134.
either by a screening test or a symptomatic com- 8. Perkins BA, Orszag A, Ngo M, Ng E, New P, Bril
plaint, requires careful consideration of the his- V.  Prediction of incident diabetic neuropathy using
the monofilament examination: a 4-year prospective
torical features, symptoms, and signs, where each study. Diabetes Care. 2010;33(7):1549–54. https://
finding revises for the clinician an estimate of doi.org/10.2337/dc09-­1835.
disease probability. Composite scores, reviewed 9. Dyck PJ, Albers JW, Andersen H, et al. Diabetic poly-
in detail in the chapter, help to remind the clini- neuropathies: update on research definition, diagnos-
tic criteria and estimation of severity. Diabetes Metab
cian of key supportive symptoms and signs for Res Rev. 2011;27(7):620–8. https://doi.org/10.1002/
diagnosis and are commonly used in cohort stud- dmrr.1226.
ies and clinical trials. Even if the clinical evalua- 10. Tesfaye S, Stevens LK, Stephenson JM, et  al.
tion is consistent with typical features of diabetic Prevalence of diabetic peripheral neuropathy and its
relation to glycaemic control and potential risk fac-
DSP, simple laboratory tests are likely to identify tors: the EURODIAB IDDM Complications Study.
the presence of other causes. It is essential that Diabetologia. 1996;39(11):1377–84. https://doi.
the clinician identify atypical features (listed in org/10.1007/s001250050586.
Table 2) or the suspicion of other causes (listed in 11. Feldman EL, Callaghan BC, Pop-Busui
R, et  al. Diabetic neuropathy. Nat Rev Dis
Table  5) to seek confirmatory tests and expert Primers. 2019;5(1):41. https://doi.org/10.1038/
opinion through neurology specialist referral. s41572-­019-­0092-­1.
12. Andersen ST, Witte DR, Dalsgaard EM, et  al. Risk
factors for incident diabetic polyneuropathy in a
cohort with screen-detected type 2 diabetes fol-
References lowed for 13 years: ADDITION-Denmark. Diabetes
Care. 2018;41(5):1068–75. https://doi.org/10.2337/
1. Pop-Busui R, Boulton AJ, Feldman EL, et  al. dc17-­2062.
Diabetic neuropathy: a position statement by the 13. Tesfaye S, Chaturvedi N, Eaton SE, et  al. Vascular
American Diabetes Association. Diabetes Care. risk factors and diabetic neuropathy. N Engl J
2017;40(1):136–54. https://doi.org/10.2337/ Med. 2005;352(4):341–50. https://doi.org/10.1056/
dc16-­2042. NEJMoa032782.
2. Manzano GM, Giuliano LM, Nobrega JA.  A brief 14. Callaghan BC, Gao L, Li Y, et al. Diabetes and obe-
historical note on the classification of nerve fibers. sity are the main metabolic drivers of peripheral neu-
Arq Neuropsiquiatr. 2008;66(1):117–9. https://doi. ropathy. Ann Clin Transl Neurol. 2018;5(4):397–405.
org/10.1590/s0004-­282x2008000100033. https://doi.org/10.1002/acn3.531.
3. Carmichael J, Fadavi H, Ishibashi F, Shore AC, 15. Prabodha LBL, Sirisena ND, Dissanayake
Tavakoli M.  Advances in screening, early diagnosis VHW.  Susceptible and prognostic genetic fac-
and accurate staging of diabetic neuropathy. Front tors associated with diabetic peripheral neu-
Endocrinol. 2021;12:671257. https://doi.org/10.3389/ ropathy: a comprehensive literature review. Int
fendo.2021.671257. J Endocrinol. 2018;2018:8641942. https://doi.
4. Wukich DK, Raspovic KM, Suder NC. Patients with org/10.1155/2018/8641942.
diabetic foot disease fear major lower-­ extremity 16. Lu B, Hu J, Wen J, et  al. Determination of periph-
amputation more than death. Foot Ankle Spec. eral neuropathy prevalence and associated factors
2018;11(1):17–21. (comparative study) (in eng). in Chinese subjects with diabetes and pre-diabetes  -
https://doi.org/10.1177/1938640017694722. ShangHai Diabetic neuRopathy Epidemiology and
5. Gregg EW, Li Y, Wang J, et al. Changes in diabetes-­ Molecular Genetics Study (SH-DREAMS). PLoS
related complications in the United States, 1990-2010. One. 2013;8(4):e61053. https://doi.org/10.1371/jour-
N Engl J Med. 2014;370(16):1514–23. (Research nal.pone.0061053.
support, U.S.  Gov’t, P.H.S.) (in eng). https://doi. 17. Kirthi V, Perumbalath A, Brown E, et al. Prevalence
org/10.1056/NEJMoa1310799. of peripheral neuropathy in pre-diabetes: a systematic
6. Geiss LS, Li Y, Hora I, Albright A, Rolka D, Gregg review. BMJ Open Diabetes Res Care. 2021;9(1):40.
EW.  Resurgence of diabetes-related nontraumatic https://doi.org/10.1136/bmjdrc-­2020-­002040.
Clinical Diagnosis of Diabetic Peripheral Neuropathy 89

18. Kraiwong R, Vongsirinavarat M, Hiengkaew V, von Lentz SI, Tavakoli M, Boulton AJM, Efron N, Malik
Heideken WP.  Effect of sensory impairment on bal- RA. Corneal confocal microscopy predicts the devel-
ance performance and lower limb muscle strength opment of diabetic neuropathy: a longitudinal diag-
in older adults with type 2 diabetes. Ann Rehabil nostic multinational consortium study. Diabetes Care.
Med. 2019;43(4):497–508. https://doi.org/10.5535/ 2021;44(9):2107–14.
arm.2019.43.4.497. 31. Kirk JK, Hunter JC, Mihalko SL, Danhauer SC,
19. Callaghan BC, Price RS, Feldman EL. Distal symmet- Shumaker SA.  Perspectives of pain in patients with
ric polyneuropathy in 2020. JAMA. 2020;324(1):90– type 2 diabetes. Expert Rev Endocrinol Metab.
1. https://doi.org/10.1001/jama.2020.0700. 2019;14(3):215–9. https://doi.org/10.1080/17446651
20. Maltese G, Tan SV, Bruno E, Brackenridge A, Thomas .2019.1592674.
S. Peripheral neuropathy in diabetes: it’s not always 32. Bai JW, Lovblom LE, Cardinez M, et al. Neuropathy
what it looks like. Diabet Med. 2018;35(10):1457–9. and presence of emotional distress and depres-
https://doi.org/10.1111/dme.13701. sion in longstanding diabetes: results from the
21. Martin CL, Albers JW, Pop-Busui R. Neuropathy and Canadian study of longevity in type 1 diabetes. J
related findings in the diabetes control and complica- Diabetes Complicat. 2017;31(8):1318–24. https://doi.
tions trial/epidemiology of diabetes interventions and org/10.1016/j.jdiacomp.2017.05.002.
complications study. Diabetes Care. 2014;37(1):31–8. 33. Callaghan BC, Cheng HT, Stables CL, Smith AL,
https://doi.org/10.2337/dc13-­2114. Feldman EL.  Diabetic neuropathy: clinical mani-
22. Scott IA, Greenberg PB, Poole PJ.  Cautionary tales festations and current treatments. Lancet Neurol.
in the clinical interpretation of studies of diagnostic 2012;11(6):521–34. https://doi.org/10.1016/
tests. Intern Med J. 2008;38(2):120–9. https://doi. S1474-­4422(12)70065-­0.
org/10.1111/j.1445-­5994.2007.01436.x. 34. Bracewell N, Game F, Jeffcoate W, Scammell
23. Dobrow MJ, Hagens V, Chafe R, Sullivan T, BE. Clinical evaluation of a new device in the assess-
Rabeneck L.  Consolidated principles for screening ment of peripheral sensory neuropathy in diabe-
based on a systematic review and consensus process. tes. Diabet Med. 2012;29(12):1553–5. https://doi.
CMAJ. 2018;190(14):422–9. https://doi.org/10.1503/ org/10.1111/j.1464-­5491.2012.03729.x.
cmaj.171154. 35. Nather A, Keng Lin W, Aziz Z, Ong C, McFeng
24. Bril V, Breiner A, Perkins BA, Zochodne D, B.  Assessment of sensory neuropathy in patients
Diabetes Canada Clinical Practice Guidelines Expert with diabetic foot problems. Diabetes Foot Ankle.
C.  Neuropathy. Can J Diabetes. 2018;42(1):217–21. 2011;2:6367. https://doi.org/10.3402/dfa.v2i0.6367.
https://doi.org/10.1016/j.jcjd.2017.10.028. 36. Gylfadottir SS, Weeracharoenkul D, Andersen
25. Perez-Panero AJ, Ruiz-Munoz M, Cuesta-Vargas ST, Niruthisard S, Suwanwalaikorn S, Jensen
AI, Gonzalez-Sanchez M.  Prevention, assess- TS.  Painful and non-painful diabetic polyneuropa-
ment, diagnosis and management of diabetic foot thy: clinical characteristics and diagnostic issues. J
based on clinical practice guidelines: a systematic Diabetes Investig. 2019;10(5):1148–57. https://doi.
review. Medicine. 2019;98(35):e16877. https://doi. org/10.1111/jdi.13105.
org/10.1097/MD.0000000000016877. 37. Dyck PJ, Overland CJ, Low PA, et al. Signs and symp-
26. Embil JM, Albalawi Z, Bowering K, Trepman E, toms versus nerve conduction studies to diagnose
Diabetes Canada Clinical Practice Guidelines Expert diabetic sensorimotor polyneuropathy: Cl vs. NPhys
C. Foot care. Can J Diabetes. 2018;42(1):S222–7. (In trial. Muscle Nerve. 2010;42(2):157–64. https://doi.
eng). https://doi.org/10.1016/j.jcjd.2017.10.020. org/10.1002/mus.21661.
27. Sharma S, Kerry C, Atkins H, Rayman G.  The 38. Zografou I, Iliadis F, Sambanis C, Didangelos
Ipswich Touch Test: a simple and novel method to T. Validation of neuropad in the assessment of periph-
screen patients with diabetes at home for increased eral diabetic neuropathy in patients with diabetes
risk of foot ulceration. Diabet Med. 2014;31(9):1100– mellitus versus the michigan neuropathy screen-
3. (Randomized Controlled Trial Research Support, ing instrument, 10g monofilament application and
Non-U.S. Gov’t Validation Studies) (In eng). https:// biothesiometer measurement. Curr Vasc Pharmacol.
doi.org/10.1111/dme.12450. 2020;18(5):517–22. https://doi.org/10.2174/1570161
28. Kanji JN, Anglin RE, Hunt DL, Panju A.  Does this 117666190723155324.
patient with diabetes have large-fiber peripheral neu- 39. Rodriguez-Sanchez B, Pena-Longobardo LM,
ropathy? JAMA. 2010;303(15):1526–32. https://doi. Sinclair AJ.  Cost-effectiveness analysis of the
org/10.1001/jama.2010.428. Neuropad device as a screening tool for early dia-
29. Perkins BA, Olaleye D, Zinman B, Bril V.  Simple betic peripheral neuropathy. Eur J Health Econ.
screening tests for peripheral neuropathy in the diabe- 2020;21(3):335–49. https://doi.org/10.1007/
tes clinic. Diabetes Care. 2001;24(2):250–6. https:// s10198-­019-­01134-­2.
doi.org/10.2337/diacare.24.2.250. 40. Ang L, Jaiswal M, Callaghan B, Raffel D, Pop-­
30. Bruce A, LEL P, Lewis EJH, Bril V, Ferdousi M, Busui R.  Sudomotor dysfunction as a measure of
Orszag A, Edwards K, Pritchard N, Russell A, small fiber neuropathy in type 1 diabetes. Auton
Dehghani C, Pacaud D, Romanchuk K, Mah JK, Neurosci. 2017;205:87–92. https://doi.org/10.1016/j.
Jeziorska M, Marshall A, Shtein RM, Pop-Busui R, autneu.2017.03.001.
90 B. A. Perkins and V. Bril

41. Novak P.  Electrochemical skin conductance: a sys- 52. Feldman EL, Stevens MJ, Thomas PK, Brown MB,
tematic review. Clin Auton Res. 2019;29(1):17–29. Canal N, Greene DA.  A practical two-step quanti-
https://doi.org/10.1007/s10286-­017-­0467-­x. tative clinical and electrophysiological assessment
42. Rajan S, Campagnolo M, Callaghan B, Gibbons for the diagnosis and staging of diabetic neuropa-
CH.  Sudomotor function testing by electrochemical thy. Diabetes Care. 1994;17(11):1281–9. https://doi.
skin conductance: does it really measure sudomotor org/10.2337/diacare.17.11.1281.
function? Clin Auton Res. 2019;29(1):31–9. https:// 53. Seferovic JP, Pfeffer MA, Claggett B, et  al. Three-­
doi.org/10.1007/s10286-­018-­0540-­0. question set from Michigan Neuropathy Screening
43. Vinik AI, Casellini CM, Parson HK. Electrochemical Instrument adds independent prognostic information
skin conductance to measure sudomotor function: the on cardiovascular outcomes: analysis of ALTITUDE
importance of not misinterpreting the evidence. Clin trial. Diabetologia. 2018;61(3):581–8. https://doi.
Auton Res. 2019;29(1):13–5. https://doi.org/10.1007/ org/10.1007/s00125-­017-­4485-­y.
s10286-­018-­0562-­7. 54. Mather KJ, Bebu I, Baker C, et  al. Prevalence of
44. Binns-Hall O, Selvarajah D, Sanger D, Walker J, microvascular and macrovascular disease in the gly-
Scott A, Tesfaye S.  One-stop microvascular screen- cemia reduction approaches in diabetes - a compara-
ing service: an effective model for the early detection tive effectiveness (GRADE) study cohort. Diabetes
of diabetic peripheral neuropathy and the high-risk Res Clin Pract. 2020;165:108235. https://doi.
foot. Diabet Med. 2018;35(7):887–94. https://doi. org/10.1016/j.diabres.2020.108235.
org/10.1111/dme.13630. 55. Hansen CS, Jensen TM, Jensen JS, et al. The role of
45. Shibata Y, Himeno T, Kamiya T, et  al. Validity serum methylglyoxal on diabetic peripheral and car-
and reliability of a point-of-care nerve conduc- diovascular autonomic neuropathy: the ADDITION
tion device in diabetes patients. J Diabetes Investig. Denmark study. Diabet Med. 2015;32(6):778–85.
2019;10(5):1291–8. https://doi.org/10.1111/ https://doi.org/10.1111/dme.12753.
jdi.13007. 56. Pop-Busui R, Lu J, Brooks MM, et al. Impact of gly-
46. Scarr D, Lovblom LE, Cardinez N, et al. Validity of cemic control strategies on the progression of dia-
a point-of-care nerve conduction device for polyneu- betic peripheral neuropathy in the bypass angioplasty
ropathy identification in older adults with diabetes: revascularization investigation 2 diabetes (BARI 2D)
results from the Canadian study of longevity in type cohort. Diabetes Care. 2013;36(10):3208–15. https://
1 diabetes. PLoS One. 2018;13(4):e0196647. https:// doi.org/10.2337/dc13-­0012.
doi.org/10.1371/journal.pone.0196647. 57. Barbosa M, Saavedra A, Severo M, Maier C, Carvalho
47. Lee JA, Halpern EM, Lovblom LE, Yeung E, Bril V, D.  Validation and reliability of the Portuguese ver-
Perkins BA. Reliability and validity of a point-of-care sion of the Michigan neuropathy screening instru-
sural nerve conduction device for identification of ment. Pain Pract. 2017;17(4):514–21. https://doi.
diabetic neuropathy. PLoS One. 2014;9(1):e86515. org/10.1111/papr.12479.
https://doi.org/10.1371/journal.pone.0086515. 58. Bax G, Fagherazzi C, Piarulli F, Nicolucci A, Fedele
48. Perkins BA, Orszag A, Grewal J, Ng E, Ngo M, Bril D. Reproducibility of Michigan neuropathy screening
V.  Multi-site testing with a point-of-care nerve con- instrument (MNSI). A comparison with tests using the
duction device can be used in an algorithm to diag- vibratory and thermal perception thresholds. Diabetes
nose diabetic sensorimotor polyneuropathy. Diabetes Care. 1996;19(8):904–5. https://doi.org/10.2337/
Care. 2008;31(3):522–4. https://doi.org/10.2337/ diacare.19.8.904.
dc07-­1227. 59. Bril V, Perkins BA. Validation of the Toronto clinical
49. Poulose S, Cheriyan E, Poulose A, Cheriyan R, scoring system for diabetic polyneuropathy. Diabetes
Vadakkanezath B, Ziemer P.  Usefulness of the Care. 2002;25(11):2048–52. https://doi.org/10.2337/
NC-stat DPNCheck nerve conduction test in a com- diacare.25.11.2048.
munity pharmacy as an educational tool for patients 60. Bril V, Tomioka S, Buchanan RA, Perkins
with diabetes. Can Pharm J. 2015;148(1):17–20. BA. Reliability and validity of the modified Toronto
https://doi.org/10.1177/1715163514561055. clinical neuropathy score in diabetic sensorimotor
50. Zhou X, Obuchowski NA, McClish DK.  Basic con- polyneuropathy. Diabet Med. 2009;26(3):240–6.
cepts and methods, introduction. In: Statistical meth- https://doi.org/10.1111/j.1464-­5491.2009.02667.x.
ods in diagnostic medicine. 2nd ed. Hoboken: Wiley; 61. Abraham A, Barnett C, Katzberg HD, Lovblom LE,
2011. p. 1–11. Perkins BA, Bril V. Toronto clinical neuropathy score
51. Herman WH, Pop-Busui R, Braffett BH, et  al. Use is valid for a wide spectrum of polyneuropathies. Eur
of the Michigan Neuropathy Screening Instrument J Neurol. 2018;25(3):484–90. https://doi.org/10.1111/
as a measure of distal symmetrical peripheral ene.13533.
neuropathy in type 1 diabetes: results from the 62. Lysy Z, Lovblom LE, Halpern EM, et al. Measurement
diabetes control and complications trial/epide- of cooling detection thresholds for identification of
miology of diabetes interventions and complica- diabetic sensorimotor polyneuropathy in type 1 dia-
tions. Diabet Med. 2012;29(7):937–44. https://doi. betes. PLoS One. 2014;9(9):e106995. https://doi.
org/10.1111/j.1464-­5491.2012.03644.x. org/10.1371/journal.pone.0106995.
Clinical Diagnosis of Diabetic Peripheral Neuropathy 91

63. Lovblom LE, Halpern EM, Wu T, et al. In vivo corneal BMC Neurol. 2021;21(1):70. https://doi.org/10.1186/
confocal microscopy and prediction of future-incident s12883-­021-­02094-­y.
neuropathy in type 1 diabetes: a preliminary longitu- 75. Ziegler D, Edmundson S, Gurieva I, Mankovsky B,
dinal analysis. Can J Diabetes. 2015;39(5):390–7. Papanas N, Strokov I. Predictors of response to treat-
https://doi.org/10.1016/j.jcjd.2015.02.006. ment with actovegin for 6 months in patients with
64. Lewis EJH, Perkins BA, Lovblom LE, Bazinet RP, type 2 diabetes and symptomatic polyneuropathy. J
Wolever TMS, Bril V. Effect of omega-3 supplemen- Diabetes Complicat. 2017;31(7):1181–7. https://doi.
tation on neuropathy in type 1 diabetes: A 12-month org/10.1016/j.jdiacomp.2017.03.012.
pilot trial. Neurology. 2017;88(24):2294–301. https:// 76. Ziegler D, Low PA, Freeman R, Tritschler H, Vinik
doi.org/10.1212/WNL.0000000000004033. AI.  Predictors of improvement and progression of
65. Sekiguchi K, Kohara N, Baba M, et al. Aldose reduc- diabetic polyneuropathy following treatment with
tase inhibitor ranirestat significantly improves nerve alpha-lipoic acid for 4 years in the NATHAN 1 trial.
conduction velocity in diabetic polyneuropathy: a J Diabetes Complicat. 2016;30(2):350–6. https://doi.
randomized double-blind placebo-controlled study org/10.1016/j.jdiacomp.2015.10.018.
in Japan. J Diabetes Investig. 2019;10(2):466–74. 77. Tesfaye S, Tandan R, Bastyr EJ, et  al. Factors that
https://doi.org/10.1111/jdi.12890. impact symptomatic diabetic peripheral neuropathy
66. Zinman LH, Ngo M, Ng ET, Nwe KT, Gogov S, Bril in placebo-administered patients from two 1-year
V. Low-intensity laser therapy for painful symptoms clinical trials. Diabetes Care. 2007;30(10):2626–32.
of diabetic sensorimotor polyneuropathy: a controlled https://doi.org/10.2337/dc07-­0608.
trial. Diabetes Care. 2004;27(4):921–4. https://doi. 78. Singleton JR, Bixby B, Russell JW, et  al. The
org/10.2337/diacare.27.4.921. Utah Early Neuropathy Scale: a sensitive clinical
67. Abraham A, Katzberg HD, Lovblom LE, Perkins scale for early sensory predominant neuropathy. J
BA, Bril V.  Uric acid levels correlate with sensory Peripher Nerv Syst. 2008;13(3):218–27. https://doi.
nerve function in healthy subjects. Can J Neurol org/10.1111/j.1529-­8027.2008.00180.x.
Sci. 2019;46(3):337–41. https://doi.org/10.1017/ 79. Boger MS, Hulgan T, Haas DW, et  al. Measures
cjn.2019.9. of small-fiber neuropathy in HIV infection.
68. Dunnigan SK, Ebadi H, Breiner A, et al. The charac- Auton Neurosci. 2012;169(1):56–61. https://doi.
teristics of chronic inflammatory demyelinating poly- org/10.1016/j.autneu.2012.04.001.
neuropathy in patients with and without diabetes–an 80. Andreasson M, Lagali N, Badian RA, et  al.
observational study. PLoS One. 2014;9(2):e89344. Parkinson’s disease with restless legs syndrome-
https://doi.org/10.1371/journal.pone.0089344. ­an in  vivo corneal confocal microscopy study. NPJ
69. Ghavami H, Radfar M, Soheily S, Shamsi SA, Parkinsons Dis. 2021;7(1):4. https://doi.org/10.1038/
Khalkhali HR.  Effect of lifestyle interventions on s41531-­020-­00148-­5.
diabetic peripheral neuropathy in patients with type 81. Fernandez-Torres R, Ruiz-Munoz M, Perez-­
2 diabetes, result of a randomized clinical trial. Panero AJ, Garcia-Romero JC, Gonzalez-Sanchez
Agri. 2018;30(4):165–70. https://doi.org/10.5505/ M.  Clinician assessment tools for patients with dia-
agri.2018.45477. betic foot disease: a systematic review. J Clin Med.
70. Wahren J, Foyt H, Daniels M, Arezzo JC.  Long-­ 2020;9(5):487. https://doi.org/10.3390/jcm9051487.
acting C-peptide and neuropathy in type 1 dia- 82. Smith AG, Singleton JR.  Obesity and hyperlipid-
betes: a 12-month clinical trial. Diabetes Care. emia are risk factors for early diabetic neuropathy. J
2016;39(4):596–602. https://doi.org/10.2337/ Diabetes Complicat. 2013;27(5):436–42. https://doi.
dc15-­2068. org/10.1016/j.jdiacomp.2013.04.003.
71. Bril V.  NIS-LL: the primary measurement scale for 83. Smith AG, Lessard M, Reyna S, Doudova M,
clinical trial endpoints in diabetic peripheral neuropa- Singleton JR.  The diagnostic utility of Sudoscan
thy. Eur Neurol. 1999;41(Suppl 1):8–13. https://doi. for distal symmetric peripheral neuropathy. J
org/10.1159/000052074. Diabetes Complicat. 2014;28(4):511–6. https://doi.
72. Dyck PJ, Davies JL, Litchy WJ, O'Brien org/10.1016/j.jdiacomp.2014.02.013.
PC. Longitudinal assessment of diabetic polyneurop- 84. Gonzalez-Duarte A, Lem M, Diaz-Diaz E, Castillo
athy using a composite score in the rochester diabetic C, Cardenas-Soto K.  The efficacy of pregabalin in
neuropathy study cohort. Neurology. 1997;49(1):229– the treatment of prediabetic neuropathic pain. Clin J
39. https://doi.org/10.1212/wnl.49.1.229. Pain. 2016;32(11):927–32. https://doi.org/10.1097/
73. Zilliox LA, Ruby SK, Singh S, Zhan M, Russell AJP.0000000000000339.
JW.  Clinical neuropathy scales in neuropathy 85. Young MJ, Boulton AJ, MacLeod AF, Williams DR,
associated with impaired glucose tolerance. J Sonksen PH.  A multicentre study of the prevalence
Diabetes Complicat. 2015;29(3):372–7. https://doi. of diabetic peripheral neuropathy in the United
org/10.1016/j.jdiacomp.2015.01.011. Kingdom hospital clinic population. Diabetologia.
74. Lin X, Yarlas A, Vera-Llonch M, et al. Rate of neu- 1993;36(2):150–4. https://doi.org/10.1007/
ropathic progression in hereditary transthyretin amy- BF00400697.
loidosis with polyneuropathy and other peripheral 86. Meijer JW, Smit AJ, Sonderen EV, Groothoff
neuropathies: a systematic review and meta-analysis. JW, Eisma WH, Links TP.  Symptom scoring sys-
92 B. A. Perkins and V. Bril

tems to diagnose distal polyneuropathy in dia- 91. Pham H, Armstrong DG, Harvey C, Harkless LB,
betes: the Diabetic Neuropathy Symptom score. Giurini JM, Veves A.  Screening techniques to iden-
Diabet Med. 2002;19(11):962–5. https://doi. tify people at high risk for diabetic foot ulcer-
org/10.1046/j.1464-­5491.2002.00819.x. ation: a prospective multicenter trial. Diabetes
87. Asad A, Hameed MA, Khan UA, Ahmed N, Butt Care. 2000;23(5):606–11. https://doi.org/10.2337/
MU. Reliability of the neurological scores for assess- diacare.23.5.606.
ment of sensorimotor neuropathy in type 2 diabetics. 92. Dhage S, Ferdousi M, Adam S, et al. Corneal confocal
J Pak Med Assoc. 2010;60(3):166–70. microscopy identifies small fibre damage and progres-
88. Jia WP, Shen Q, Bao YQ, Lu JX, Li M, Xiang sion of diabetic neuropathy. Sci Rep. 2021;11(1):1859.
KS. Evaluation of the four simple methods in the diag- https://doi.org/10.1038/s41598-­021-­81302-­8.
nosis of diabetic peripheral neuropathy. Zhonghua Yi 93. Ferdousi M, Kalteniece A, Azmi S, et al. Corneal con-
Xue Za Zhi. 2006;86(38):2707–10. focal microscopy compared with quantitative sensory
89. Abbott CA, Carrington AL, Ashe H, et  al. The testing and nerve conduction for diagnosing and strat-
North-West Diabetes Foot Care Study: inci- ifying the severity of diabetic peripheral neuropathy.
dence of, and risk factors for, new diabetic foot BMJ Open Diabetes Res Care. 2020;8(2):e001801.
ulceration in a community-based patient cohort. https://doi.org/10.1136/bmjdrc-­2020-­001801.
Diabet Med. 2002;19(5):377–84. https://doi. 94. Cornblath DR, Chaudhry V, Carter K, et  al.
org/10.1046/j.1464-­5491.2002.00698.x. Total neuropathy score: validation and reliability
90. Weintrob N, Amitay I, Lilos P, Shalitin S, Lazar L, study. Neurology. 1999;53(8):1660–4. https://doi.
Josefsberg Z.  Bedside neuropathy disability score org/10.1212/wnl.53.8.1660.
compared to quantitative sensory testing for mea- 95. Menten J, Lesaffre E. A general framework for com-
surement of diabetic neuropathy in children, ado- parative Bayesian meta-analysis of diagnostic studies.
lescents, and young adults with type 1 diabetes. J BMC Med Res Methodol. 2015;15:70. https://doi.
Diabetes Complicat. 2007;21(1):13–9. https://doi. org/10.1186/s12874-­015-­0061-­7.
org/10.1016/j.jdiacomp.2005.11.002.
Diagnostic Techniques for Diabetic
Peripheral Neuropathy

Long Davalos, Amro Stino, and A. Gordon Smith

1 Introduction tions where diagnostic testing plays an important


role. Diagnostic tests can be used to confirm the
The most common peripheral neuropathy associ- suspected diagnosis for clinical or research pur-
ated with both type 1 (T1D) and type 2 diabetes poses and as surrogate measures of disease sever-
(T2D) is a distal symmetric polyneuropathy ity in natural history or therapeutic trials. The
(DPN). DPN is characterized by length-­ clinical spectrum of DPN is reflected in the vari-
dependent sensory loss predominantly affecting ety of diagnostic tests available that evaluate dif-
the distal lower extremities with minimal weak- ferent nerve fiber classes and functions.
ness. There are several phenotypic variants of Appropriate application of these tests is based on
DPN, ranging from painful sensory neuropathy the specific clinical phenotype and context of use
(sometimes in the setting of preferential injury to (clinical diagnosis, epidemiological studies, clin-
small unmyelinated axons), nonpainful neuropa- ical trials, etc.).
thy with sensory symptoms, and asymptomatic Nerve conduction studies (NCS) are the most
DPN, which can present with an insensate foot, widely used diagnostic modality, and assess
often in the setting of foot ulceration. Diagnosis large, myelinated motor, and sensory nerve fibers
of DPN is largely based on a combination of through electrical stimulation of a peripheral
symptoms and signs, with signs conveying nerve with recording of the resulting nerve or
greater diagnostic certainty, as summarized in muscle response. The Toronto consensus criteria
chapter “Clinical Diagnosis of Diabetic require abnormalities of NCS to establish a diag-
Peripheral Neuropathy” by Dr. Bruce Perkins. nosis of “confirmed DPN,” although for clinical
However, there are clinical and research situa- diagnostic purposes, NCS are not necessary for
many patients with signs and symptoms of DPN
(Table 1) [1, 2]. Although NCS objectively quan-
L. Davalos tify large myelinated fiber function, they do not
Department of Neurology and Rehabilitation
Medicine, University of Cincinnati, assess small fibers [3]. A diagnosis of confirmed
Cincinnati, OH, USA small fiber neuropathy (SFN) requires an abnor-
A. Stino mality of a validated measure of small fiber func-
Department of Neurology, University of Michigan tion [4]. Skin biopsy for measurement of
Medicine, Ann Arbor, MI, USA intraepidermal nerve fiber density (IENFD) is the
A. G. Smith (*) best validated measure of small fiber function but
Department of Neurology, Virginia Commonwealth several other techniques are available with vary-
University, Richmond, VA, USA ing degrees of validation supporting their use,
e-mail: Gordon.Smith@vcuhealth.org

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 93


S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_6
94 L. Davalos et al.

Table 1  Toronto consensus criteria. Definitions of mini- Wiehl Germany), a simple measure of sudomotor
mal criteria for typical diabetic polyneuropathy
function.
Diagnostic This chapter reviews each of these testing
category Definition
modalities including their validity and reliability,
Possible The presence of any symptom or sign of
DSPN DSPN. Symptoms include decreased and current utility in clinical and research settings.
sensation or positive neuropathic
sensory symptoms predominantly in the
toes, feet, or legs. Signs include distal 2 Nerve Conduction Studies
symmetric decrease in sensation or
unequivocally decreased or absent ankle
reflexes NCS are considered the gold standard for DPN
Probable The presence of a combination of diagnostic confirmation and are routinely used in
DSPN symptoms and signs of neuropathy clinical research settings to assess disease pro-
including any two or more of the
gression or response to therapy. In the clinical
following: neuropathic symptoms,
decreased distal sensation, or setting, NCS can be used as a quantitative confir-
unequivocally decreased or absent ankle matory test, especially when a patient presents
reflexes with features that are atypical for DPN, such as
Confirmed The presence of an abnormality of NCS acute or subacute onset, non-length dependence,
DSPN and a symptom or symptoms or a sign
or signs of neuropathy. If NCS is motor predominance or significant asymmetry of
normal, a validated measure of SFN neuropathic signs or symptoms [9]. While NCS
may be used have a high degree of reproducibility, particularly
Subclinical The presence of no signs or symptoms nerve conduction velocity, they require signifi-
DSPN of neuropathy are confirmed with
cant technical skill and are operator dependent.
abnormal NCS or a validated measure
of SFN
DSPN distal symmetric polyneuropathy, NCS nerve con-
duction studies, SFN small fiber neuropathy 2.1 Basic Principles and NCS
Abnormalities in DPN

including corneal confocal microscopy and mea- NCS involve electrically stimulating a nerve and
sures of sudomotor (sweat) function, which is recording the resulting action potential. Sensory
subserved by small diameter unmyelinated axons nerve action potentials (SNAPs) are recorded by
that are frequently injured in patients with placing surface electrodes over a cutaneous nerve
somatic SFN [5–8]. Small and large fiber func- (Fig.  1). Compound muscle action potentials
tion can be assessed using quantitative sensory (CMAPs) are obtained by placing surface elec-
testing for thermal and pain or vibration sensa- trodes over a muscle supplied by the nerve. The
tion respectively. Each of these techniques amplitude of the response is reflective of the
requires special equipment and technical exper- number of axons contributing to the summated
tise and, in the case of IENFD, access to a waveform. The time it takes for the response to
­qualified laboratory. While each may be applied occur (distal latency) and the conduction velocity
in varying research settings, from a clinical per- (CV) reflect the function of peripheral nerve
spective, there is interest in development of con- myelination. Loss of amplitude suggests axonal
venient and well-tolerated point of care loss or dysfunction, whereas slowing of CV or
techniques that can be used to confirm DPN or latency prolongation usually implies demyelin-
for disease screening purposes. These include ation [10]. F-wave latencies also asses nerve
several quantitative sensory testing (QST) function and represent the motor conduction time
devices (vibrameter, neurometer, and thermoaes- to and from the spinal cord along the entirety of
thesiometer), point of care NCS devices (DPN the motor nerve axon. Therefore, it assesses the
Check®, NeuroMetrix, Massachusetts USA), and most proximal nerve segment and the entire
Neuropad® (TRIGOcare International GmbH, length of a motor nerve.
Diagnostic Techniques for Diabetic Peripheral Neuropathy 95

tionally treated group decreased by 0.56 and 0.54


m/s/year, respectively, over 5 years [14]. In a pro-
spective 8-year study of 45 T1D patients, a 1%
rise in HbA1c was associated with a 1.3 meter per
second decrease in maximal nerve CV [15].
Furthermore, in patients with T2D, a lower rate of
decline was observed in a 10-year natural history
study of 133 patients with newly diagnosed T2D,
in which NCV deteriorated by 0.39 m/s/year in
the sural and 0.3 m/s/year in the peroneal nerves
[13]. In regard to the F-wave, prolongation of the
F-wave latencies has shown to be very sensitive
for DPN as well as highly reproducible [16, 17].
The amplitude reduction of the nerve action
potentials with disease progression is a reflection of
the axonal degeneration and loss of nerve fibers
observed in DPN, which is the hallmark of nerve
injury in this condition [18, 19]. The degree of CV
slowing in DPN is not as prominent as the slowing
seen in primary demyelinating neuropathies such as
acute or chronic inflammatory demyelinating poly-
radiculoneuropathy. These changes could reflect
Fig. 1  Recording electrodes are placed over the first digit
mild demyelination or a loss of nodal ion channels
and an electrical stimulation is applied to the median
nerve at the wrist (top panel). The resulting sensory nerve without a structural alteration, as is shown in some
action potential (SNAP) is illustrated below. The ampli- studies [20]. The prolonged F-wave latencies may
tude of the response is proportionate to the number of indicate more widespread nerve involvement. Some
large diameter sensory axons, and conduction velocity to
pathological DPN studies indicate distally promi-
myelin function
nent but diffuse involvement of peripheral nerves
[21, 22], while other studies show patchy fiber
Patients with DPN have reduced SNAP ampli- degeneration with fascicular sprouting that begins
tudes, often with mildly slowed motor nerve con- proximally and extends distally [18].
duction velocity, and mildly prolonged minimal
F-wave latencies [11]. DPN is a length-­dependent
process involving the longest nerves, thus find- 2.2 Clinical Utility of NCS in DPN
ings are most prominent in the distal lower Diagnosis
extremities. A reduction of 1 μV in the sural nerve
SNAP amplitude associates with a decrease of The consensus regarding the diagnosis of DPN
approximately 150 fibers/mm2, while a mean has changed throughout the years. The 1998 San
reduction of 1mV of the ulnar, peroneal, and tibial Antonio Conference on Diabetic Neuropathy
nerves CMAP amplitudes associates with a den- [23], and then the American Academy of
sity loss of 200 fibers/mm2 [12]. SNAP ampli- Neurology (AAN), American Association of
tudes decline over time, with one study of patients Electrodiagnostic Medicine (AAEM), and
with DPN associated with T2D experiencing a American Academy of Physical Medicine and
5% loss of sural SNAP amplitude per year [13]. Rehabilitation (AAPM&R) in 2005 [24] pro-
Nerve CV also decreases with disease progres- posed criteria for DPN that included abnormali-
sion. In the Diabetic Control and Complications ties on NCS.  The Toronto consensus criteria
Trial, which assessed T1D patients, the sural sen- (2010) defined confirmed DPN symptoms and
sory and peroneal motor nerve CV in the conven- signs in addition to abnormal NCS [1].
96 L. Davalos et al.

The AAN, AANEM, and AAPM&R recom- show that NCS is an objective and reliable test
mended a clinical research protocol for NCS in for the diagnosis of DPN. In comparison to other
2005 that proposed the specific studies necessary, methods such as clinical examination or QST,
including unilateral examination of sural, ulnar NCS has lower coefficient of variation on repeat
and median sensory nerves, and peroneal, tibial, testing [34–36]. Furthermore, NCS abnormalities
median and ulnar motor nerves with F waves. An seem to correlate with nerve morphology [37]
abbreviated protocol was also proposed, more and clinical severity of disease [38]. These attri-
practical for the clinical setting, which included a butes make NCS a reliable and standardized tool
sural sensory and peroneal motor nerve in one to measure nerve function. However, there are
lower extremity. The minimum case definition cri- some limitations to consider when utilizing NCS
terion for electrodiagnostic confirmation was an for diagnostic confirmation. NCS cannot detect
abnormality (≥99th or ≤1st percentile) of any small fiber nerve injury, which may be the earli-
attribute of nerve conduction in two separate est detectable sign in DPN, particularly in patients
nerves, one of which had to be the sural nerve [24]. with prominent neuropathic pain [3]. While this
In 2011, the Nerve Conduction Criteria Study is a relevant limitation for studies focusing on
reported that the six most sensitive attributes of small fiber predominant neuropathy, most
NCS for diagnosing subclinical DPN were pero- patients with symptomatic DPN experience pro-
neal motor CV, sural sensory amplitude, tibial gressive loss both small unmyelinated and large
motor CV, ulnar motor CV, tibial, and ulnar F-wave myelinated axons [39]. Furthermore, the overall
latencies. This study evaluated eight different elec- population normative ranges for NCS parameters
trodiagnostic criteria and concluded that utilizing may overlap with abnormal values in DPN unless
the “≥1 abnormal attributes (≤1st/≥99th or age, gender, height, and body mass index are
≤2.5st/≥97.5th) in ≥2 separate nerves” criteria considered [40]. While intraobserver reliability is
was acceptable for clinical practice [25]. high, unless meticulous attention is paid to stan-
These criteria provide general guidance to dardized protocols and training, interobserver
practicing clinicians, but they are largely intended reliability can be unacceptably low [41, 42]. This
for research settings. While NCS are objective issue is a concern, especially for multicenter ther-
and reliable, they have some pitfalls that should apeutic trials, where even very small interob-
be taken into account in the clinical setting. NCS server differences can decrease the statistical
require a referral to a neurophysiology lab with power of a study to detect a meaningful change in
trained examiners. The procedure is uncomfort- disease course.
able, and is a significant driver of health care
costs attributable to neuropathy evaluation [26]. In order to use NCS as a quantitative diagnos-
NCS rarely alter the care of patients with DPN tic method and overcome its limitations, it is nec-
[27], and routine use for diagnostic confirmation essary to standardize the common reference
of typical DPN is not recommended. NCS should values, percentile abnormalities, and the exact
be performed in situations where the clinical fea- placement of stimulating and recording elec-
tures are atypical (motor greater than sensory trodes among EMG laboratories. Implementing
neuropathy, rapid onset, or asymmetrical presen- these measures and encouraging the same clini-
tation), the diagnosis is unclear, or a different eti- cal neurophysiologist to perform all serial NCS
ology is suspected [28]. on a given subject improves the interobserver
reliability and the accuracy of NCS as a diagnos-
tic and research outcome measure tool [42].
2.3 Research Applications for NCS
Defining DPN Using NCS  The definition of
Advantages and Limitations of NCS  Different DPN among studies varies considerably. Some
DPN cohorts [19, 29, 30] and population-based define DPN only clinically, while others utilize
studies of healthy and diabetic subjects [31–33] NCS, QST or a combination. For this reason,
Diagnostic Techniques for Diabetic Peripheral Neuropathy 97

there is great variability in the reported preva- NCS as an Endpoint Measure in Clinical
lence of DPN (8–50%) [13, 43, 44]. The Toronto Trials and Natural History Studies  Multiple
consensus advocates the use of NCS and clinical clinical trials have used NCS as the primary
abnormalities, and defines confirmed typical outcome to assess treatment efficacy and have
DPN as the presence of an abnormality of NCS failed to meet this primary endpoint [48–51]. In
along with symptom(s)/sign(s) of neuropathy [1]. this regard, there are several findings to suggest
This definition is simple and can be used to estab- that using NCS as an isolated endpoint measure
lish the diagnosis of DPN with a high degree of is not ideal. NCS cannot detect small fiber
certainty, although for some epidemiological nerve function [52], suffer from high interob-
studies where NCS are impractical due to access server variability, and the clinical meaning of
or cost, clinical criteria alone suffice (signs and modest changes in nerve conduction velocity
symptoms—“probable DPN”). Composite scores (the most commonly used single attribute as a
of neurologic signs and NCS attributes can fur- study endpoint measures) has not been well
ther increase the sensitivity and specificity of the defined. The American Diabetes Association
diagnosis [31, 45]. There is not a formal consen- recommends using validated clinical instru-
sus regarding specific NCS criteria for DPN. An ments combined, with electrophysiology; mea-
abnormality of one or more attributes (exceeding sures of small fiber damage and repair (e.g.
the normal limits between the 1st and 99th per- intraepidermal nerve fiber density), and objec-
centiles; variables, such as age, height, and tem- tive measures of patient function in the design
perature, should be considered when developing of DPN trials [28]. Utilizing composite sum
the reference range and interpreting the results) scores instead of individual attributes can
in two or more separate nerves is an acceptable increase the sensitivity of NCS as an endpoint
criterion for the electrodiagnostic definition of measure [25, 45].
DPN [46, 47].

Defining subclinical DPN is more compli- Point of Care NCS Devices  One strategy to
cated. For research purposes, the Toronto consen- overcome the technical challenges of NCS has
sus criteria defined subclinical DPN as the been to develop point of care devices capable or
presence of no signs or symptoms of neuropathy assessing specific NCS parameters (reliably
confirmed with abnormal NCS or a validated when used by individuals with minimal training).
measure of SFN [1]. However, it did not specify Point of care devices typically stimulate the sural
the minimum electrodiagnostic criteria needed, nerve at the level of the ankle while recording the
which is crucial for defining this group. The SNAP amplitude and conduction velocity using a
Nerve Conduction Study evaluated eight differ- biosensor that covers a wide surface area in the
ent criteria (criteria 1–4 evaluated individual proximal lower limb. One device demonstrated
NCS attributes and criteria 5–8 composite sum high sensitivity (95%) and acceptable specificity
scores of NCS attributes) and concluded that all (71%) for detecting DPN, and demonstrated
four composite sum scores of normal deviates good intrarater and interrater reproducibility. The
(from percentiles) performed excellently in the SNAP amplitude was similar to that obtained
healthy subject and diabetic patient cohorts, par- with traditional NCS, however, the device over-
ticularly criteria 5 and 6, which included the estimated the sural CV [53]. Overall, despite
composite sum score of two nerve attributes (e.g., these limitations, point of care NCS devices are a
peroneal CV and sural amplitude). From the indi- promising diagnostic tool that are fast, inexpen-
vidual NCS attribute criteria, Criterion 2 (≥1 sive, and easy to use. Further studies are required
abnormal attribute in 2 separate nerves) was also to evaluate performance in longitudinal clinical
acceptable [25]. trials.
98 L. Davalos et al.

3 Skin Biopsy a
with Measurement
of Intraepidermal Nerve
Fiber Density (IENFD)

Skin biopsy for IENFD measurement is a well


validated and reproducible measure of small
unmyelinated axons. Given that NCS do not eval- b
uate this fiber class, there has been a need for a
validated measure of small fiber function.
Langerhans first recognized epidermal nerve
fibers in the 1860s [54], but the existence of epi-
dermal innervation was controversial for much of
the twentieth century since epidermal nerves were
difficult to visualize using standardized histo- c
chemical techniques. Early studies focused on the
density and distribution of Meissner’s corpuscles,
but this technique was of little diagnostic utility
[55]. The availability of an antibody to protein
gene product (PGP) 9.5, a neuronal form of ubiq-
uitin hydrolase, rapidly led to sensitive immuno-
histochemical techniques to visualize nerve fibers
in the skin [56]. This technique has facilitated Fig. 2  Skin biopsies taken from the proximal (a) and dis-
tal thigh (b) and distal leg (c) of a patient with diabetic
widespread study of cutaneous innervation for neuropathy stained with PGP 9.5 demonstrate absent
both clinical and research applications (Fig. 2). IENFD at the distal site with reduced numbers of IENFD
at the distal thigh, where there are frequent axonal
swellings
3.1 Skin Anatomy and Epidermal
Innervation a b

Skin areas are classified as glabrous (hairless) or


nonglabrous. Glabrous skin is located on the
soles, palms, and fingertips, and has more sweat
glands (Fig.  3) and cutaneous sensory organs
dedicated to touch, position, and vibration sensa-
tion. On the other hand, nonglabrous skin has a
greater number of piloerector muscles and hair
follicles, which are supplied by small autonomic
nerves [57]. Both skin areas have abundant epi-
dermal nerves in normal individuals. The epider-
mis is the outermost layer of the skin, which
Fig. 3  A skin biopsy stained with PGP9.5 from a control
contains keratinocytes. The dermis harbors cuta- subject demonstrates a normally innervated sweat gland
neous sensory organs, hair follicles, blood and (a). A biopsy from a patient with diabetic neuropathy
lymphatic vessels, and sweat, apocrine and seba- demonstrates a severely denervated sweat gland (b)
ceous glands. The basement membrane (dermal-­
epidermal junction) is organized as papillae loops. Sensory organs such as Meissner corpus-
(dermal undulations extending into the epider- cles and Merkel cells are located in the apex of
mis) that contain vascular plexus and capillary these papillae.
Diagnostic Techniques for Diabetic Peripheral Neuropathy 99

Epidermal innervation arises from the net- tiple antibodies with different fluorochromes to
work of nerve fibers that run beneath the base- be used (multiple structures can be visualized
ment membrane. Intraepidermal fibers (IENFs) simultaneously) and allows three-dimensional
lose their myelin sheath as they cross the dermal-­ reconstruction of skin sections, but is more
epidermal junction and run toward the skin sur- expensive and time consuming.
face between keratinocytes. A single dermal fiber IENFD is determined by counting the number
typically divides into several intraepidermal of PGP9.5-positive fibers crossing the dermal-­
branches after crossing the basement membrane epidermal junction in at least three sections and
and extends in parallel fashion to near the surface dividing this by the length of the epidermal sur-
of the skin. In healthy subjects, IENFs have a face (measured with computer software). This
simple morphology, with small bead-like vari- value is then compared with normative age-­
cosities [58], and the density of epidermal inner- matched ranges [59, 62]. Quantification of linear
vation is greater at more proximal sites on the IEFND has a high interobserver, intraobserver,
body than it is at distal locations [59, 60]. and interlaboratory agreement [63], correlates
significantly with stereological techniques of
skin nerve morphology [64, 65], and with quanti-
3.2 Selection of Biopsy Site, fication of nerve fibers per epidermal area [66].
Processing Technique, Furthermore, there is no significant variation
and Quantification of Skin with the IEFND calculated from adjacent sec-
Nerve Fibers tions of the same biopsy or in adjacent biopsies
from the same site [59]. IEFND declines with
Skin biopsy is a relatively benign procedure. A age, is lower in males, and is not influenced by
3 mm punch biopsy is obtained following injec- weight or height [67].
tion of local anesthetic. An adhesive bandage is
used to cover the biopsy site, which generally
heals through a process of granulation within 3.3 Skin Biopsy Abnormalities
7–10 days. There is a very low risk of local infec- in DPN
tion. Selection of the biopsy site depends on the
clinical context and intent. For diagnosing length-­ Patients with painful neuropathy associated with
dependent neuropathies, as in DPN, one skin diabetes often have preferential injury to small
biopsy sample is taken from the distal part of the unmyelinated axons [3]. Abnormalities in distal
leg (10  cm above the external malleolus) and leg IEFND have been reported in 47.9% of
another specimen is obtained from the lateral patients with early diabetes and 12.5% of predia-
aspect of the proximal thigh (20  cm below the betic individuals. Furthermore, using IEFND
anterior iliac spine) [61]. The tissue needs to be abnormalities as a criterion for confirmed DPN in
immediately placed in either 2% periodate-­ early diabetes detects a higher number of patients
lysine-­
paraformaldehyde (PLP) or Zamboni with neuropathy (37.5%) in comparison to NCS
solution (2% paraformaldehyde, picric acid) for (only 14.6%). Similarly, the proportion of
12–24 h at 4 °C, and is then placed in cryoprotec- patients with early diabetes with abnormal NCS
tant solution. After that, the samples can be stored is 4.2% compared to 16.7% for IENFD [3]. As
in a freezer or sent to a processing center. The diabetes progresses, studies also show concor-
biopsied tissue is cut into 50 μm sections and dant progressive reduction in IENFD that follows
stained with anti PGP 9.5 antibody, which avidly disease course. IENFD declines in patients with
stains all axons [56]. IENFD can then be ana- DPN at a rate of between 1 and 3 fibers/mm/year
lyzed via two different microscopic techniques. [68]. In regards of neuropathic severity, IENFD
The most commonly used method is standard inversely correlates with the Neurological
bright field microscopy since it is convenient, Disability Score [69]. One study reported no
inexpensive, and relatively easy to use. The other ­correlation between IENFD and the Neuropathy
method is confocal imaging, which allows mul- Symptom Score, but showed an inverse correla-
100 L. Davalos et al.

tion with pain severity [70]. Patients with painful tory studies and clinical trials. Small
DPN tend to have more severe IENFD loss com- unmyelinated axons may be particularly suscep-
pared to those without pain, particularly when tible to injury while being more capable of
there are little or no objective signs of neuropa- regeneration than large myelinated fibers [75].
thy. It is possible that IENFD loss plays an impor- These characteristics may explain why skin
tant role in the early evolution of neuropathic biopsy is more sensitive to disease progression
pain, while other mechanisms play a more rele- than NCS [76, 77].
vant role at more advanced stages [71]. The relatively uniform and superficial loca-
tion of the IENF in the epidermis allows for the
standardized assessment of nerve injury and
3.4 Clinical Application of Skin regeneration. Several models take advantage of
Biopsy the small fibers’ uniquely robust regenerative
capacity, allowing for the accurate measurement
Normative data adjusted for sex and age are avail- of collateral [78] and regenerative sprouting [52]
able for IENFD based on the typically performed in human subjects. Following removal of a con-
brightfield technique [62]. The diagnostic yield of ventional 3 mm skin biopsy, the site heals by a
skin biopsy varies based on the normative values, process of granulation and eventually becomes
imaging technique, and diagnostic gold standard re-epithelialized. The collagen plug within the
definition. In a study of 210 patients with SFN, dermis of the healing biopsy site acts as a barrier
which included 65 diabetic patients, Z-scores and to the distal ends of the transected nerves.
5th percentile cutoffs provided the highest speci- Therefore, reinnervation of the epidermis within
ficity (98 and 95%, respectively) but the lowest the original 3  mm biopsy site can only occur
sensitivity (31 and 35%, respectively) compared through sprouting of uninjured fibers surround-
with the receiver operating characteristic analysis ing the incision line. The degree of collateral
(specificity 64%, sensitivity 78%) [72]. Another sprouting can be measured by a concentric 4 or
smaller study of 58 patients with pure SFN, which 5 mm biopsy centered on the healed 3mm biopsy
included 19 diabetic patients, showed that a cut-off site. The most promising technique to induce
IENF density of ≤8.8/mm at the ankle was associ- cutaneous denervation involves application of
ated with a sensitivity of 77.2% and a specificity of capsaicin either topically as a cream or injected
79.6% in diagnosing SFN [73]. In a more recent into the epidermis [79, 80]. When applied topi-
study in patients with T1D, IENFD showed a sen- cally in an occlusive bandage, capsaicin pro-
sitivity of 61% and specificity of 80% for DPN duces a standardized injury that is reproducible
[74]. The European Federation of the Neurological and well-tolerated [52]. Studies in healthy con-
Societies and the Peripheral Nerve Society con- trol subjects and diabetic patients demonstrate
clude that IENFD is a reliable and efficient tech- that functional abnormalities in regenerative
nique to confirm SFN with a level A sprouting are present among subjects with diabe-
recommendation [67]. Skin biopsy with assess- tes with no signs or symptoms of neuropathy
ment for IENFD may be used to confirm small [52], suggesting that regenerative abnormalities
fiber predominant DPN if NCS are normal, may be an early sign of nerve dysfunction. Thus,
although a typical clinical DPN may be diagnosed the capsaicin model holds promise as a sensitive
based on clinical criteria (as is the case with NCS). means of detecting early neuropathy and evalu-
ating the efficacy of potential therapeutic inter-
ventions, early in the disease course of
3.5 Research Application of Skin DPN. Given that reinnervation occurs relatively
Biopsy rapidly, sometimes as early as 10–12 weeks, less
patients can be studied over a short period of
Skin biopsy is a powerful research tool for the time with this technique, as opposed to NCS,
study of DPN and is commonly used as a surro- which may require larger numbers of patients
gate measure of disease severity in natural his- over years.
Diagnostic Techniques for Diabetic Peripheral Neuropathy 101

4 Quantitative Sensory Testing variation of up to 150% when using the limits


method, which significantly declined when using
Quantitative sensory testing (QST) is a psycho- the levels method [86]. In contradistinction to
physical evaluation that is particularly valuable NCS, QST does not localize abnormal findings to
in DPN clinical evaluation and research as it is the peripheral nervous system, as aberrancies in
the only quantitative testing method that captures sensory perception can be traced to any site
the patient experience of sensory loss or neuro- between the skin and sensory cortex in the central
pathic pain [81]. Using pre-set automated vibra- nervous system. In addition, QST can often be
tion and thermal thresholds, QST allows for a bulky, expensive, and available only at referral
standardized application of stimulus intensity centers, limiting its widespread adoption. The
and quantification sensitivity to stimuli for both American Academy of Neurology assigned QST a
large and small fibers (vibration and thermal per- Class II rating as a diagnostic test, concluding it
ception respectively). A number of different test- was a useful tool in research studies but should not
ing platforms are available. be the sole criteria for neuropathy diagnosis [87].
QST provides relatively consistent inter and
intra rater reliability, and is now routinely used as
an endpoint in multicenter clinical trials of DPN 4.1 Point of Care QST Devices
[82]. It has proven itself useful clinically to assess
for asymptomatic DPN [83], to uncover patients at Given the challenges of using large, expensive,
risk for foot ulceration [83], and to assess disease and non-portable QST platforms, numerous
severity longitudinally [84]. In one study of 117 mobile point of care devices are available. Some
patients with painful sensory neuropathy (not lim- devices focus purely on one modality. Vibrometers
ited to diabetic patients), QST was found to have a may be used to assess large myelinated A-beta
diagnostic sensitivity of 72%, compared to 59% fibers. The simplest tool is the graduated Rydel-­
and 87.5% for quantitative sudomotor axon reflex Seiffer tuning fork, which employs a semiquanti-
testing (QSART) and IENFD, respectively [85]. tative 8 point scale (Fig.  4). A battery-operated
However, QST does suffer from inter-­assessment handheld vibrometer (VibraTip™, McCallan

a b

Fig. 4  The Rydel-Seiffer tuning fork is a semiquantita- fork are pinched and released. The triangles formed by the
tive tool to assess vibration sensation. The tuning fork is movement allow definition of the number when the sensa-
placed on the great toe (a) and the two arms of the tuning tion is no longer perceived (b)
102 L. Davalos et al.

Medical, Ottawa, Canada) may also be used to skin and current is applied and increased until it
assess vibration perception at the great toe using is felt. Once sensed, it is terminated, and
a stimulus of 128 Hz, and has been shown to decreased in increments of 0.08 mA and reap-
associate with the neuropathy disability score plied. In such a fashion, the device calculates and
[88] and demonstrated agreement with a vibra- displays the CPT, which ranges from 1 to 25 (this
tion perception threshold of >25 V, a value indi- can be done for each frequency). Values from 6 to
cating “at risk feet.” [89] The NeuroQuick™ 13 are deemed normal, with values of 1–5 (hyper-
(Schweers, Meerbusch, Germany) is a portable esthesia) and 14–25 (hypoesthesia) both being
device that assesses cold perception threshold abnormal and representing sensory neuropathy.
[90]. It features an in-built fan that emits cold air The Neurometer has been extensively studied in
at velocities of varied intensity onto the dorsum DPN. T1D subjects with essentially normal neu-
of the foot. The NeuroQuick threshold is defined ropathy symptom metrics have reduced CPT,
as the airflow at which the subject first recognizes suggesting a potential role as a screening tool for
the stimulus. The result correlates with thermal asymptomatic DPN patients [94]. T2DPN
perception thresholds on standard QST [90]. patients have higher CPT than controls when
NeuroQuick is also able to discriminate between stimulated at 2000 Hz, and CCP negatively cor-
diabetic patients with DPN, diabetic patients relates with motor and sensory nerve conduction
without DPN, and control subjects. It is more velocities [95]. CPT can distinguish patients with
sensitive at detecting DPN than thermal percep- DPN from control subjects at all three frequen-
tion threshold, making it a useful adjunctive test cies. However, reproducibility is better in con-
to diagnose early and asymptomatic DPN [90]. trols than diabetic patients [96]. In addition, one
One German study, however, found NeuroQuick Korean study cast doubt on the additive value of
to suffer from low sensitivity in identifying pain- CPT beyond conventional DPN evaluations [97].
ful SFN (irrespective of diabetes status), when Among 241 Korean patients with diabetes, CPT
compared with standard QST cold detection was higher at all frequencies among those with
threshold technique [91]. The authors concluded DPN compared to those without. However, diag-
that it was not an adequate stand-alone screen for nostic sensitivity was low. For instance, 87% of
cold hypoesthesia in patients with chronic neuro- those with reduced sensation to a 10 g monofila-
pathic pain. ment had normal CPT at 2000 Hz. This study cast
Other devices are available to assess both doubt on the additive value of CPT beyond stan-
large and small fiber function. NerveCheck™ dard clinical measures (128 Hz tuning fork, 10-g
(Phi Med Europe S.L., Barcelona, Spain) is a monofilament, and reflex sensitivity).
portable QST device that performs both vibratory
and thermal testing and has good test-retest reli-
ability and similar diagnostic accuracy to tradi- 4.2 Application of QST in Clinical
tional QST equipment [92]. The Neurometer® Trials
(Neurotron, Baltimore, MD) applies a constant
current via surface electrodes across three cur- While the diagnostic role of QST in clinical set-
rencies—2000 Hz, 250 Hz, and 5 Hz—to selec- tings is limited, and its correlation with pain
tively stimulate different nerve fiber types [93]. symptoms reported by patients is low [98], QST
The current perception threshold (CPT), the min- remains a useful research tool that holds particu-
imum intensity detectable, is used to ascertain the lar promise for predicting patient response to
presence and severity of DPN as well as the spe- neuropathic pain treatments. Numerous trials
cific fiber type involved. Each stimulus frequency have used QST metrics as outcome measures.
selects for a particular fiber subtype, such that The German Research Network on Neuropathic
2000  Hz stimulates large myelinated fibers and Pain (DFNS) has gathered QST and other pheno-
5 Hz selects for small unmyelinated fibers. Two typic data on over 1400 patients with various
electrodes coated with gel are applied to intact neuropathic pain states. The DFNS QST protocol
Diagnostic Techniques for Diabetic Peripheral Neuropathy 103

includes assessment of 13 different mechanical ing from laboratory based assessments to simple
and thermal tests, permitting a more nuanced point of care tools. Sudomotor testing is a sensi-
assessment of distinct pain phenotypes. Patients tive diagnostic tool for small fiber neuropathy
with peripheral neuropathy most commonly have caused by diabetes and other etiologies [102].
sensory loss alone (52%), with 21% having a
mixture of positive and negative sensory signs
[99]. The DFNS protocol can be used to broadly 5.1 Quantitative Sudomotor Axon
classify patients into three distinct sensory phe- Reflex Testing (QSART)
notypes: sensory loss (thermal and mechanical
sensory loss), thermal hyperalgesia (preserved QSART is a well validated reproducible measure
sensory function with heat or cold hyperalgesia), of postganglionic sudomotor fibers. Acetylcholine
and mechanical hyperalgesia (loss of thermal is iontophoresed through a central chamber of a
sensory with mechanical hyperalgesia). Patients capsule placed on the skin, which activates nerve
may also be classified into those with sensory fibers underlying the chamber. When the action
loss (the “non-irritable nociceptor phenotype) potential reaches a branch point in the nerve, it
versus those with preserved small fiber function propagates orthodromically along the other
but hyperalgesia (the “irritable nociceptor” pat- branch, stimulating sweat glands. The QSART
tern) [100]. device measures sweat production in the area
There is evidence to suggest that specific pain under the capsule surrounding the central cham-
phenotypes may predict treatment response. The ber. QSART is typically performed at four
COMBO-DN study, which compared duloxetine locations.
and pregabalin for painful DPN, demonstrated Among 100 patients with suspected small
that these agents have different effects on specific fiber neuropathy, the diagnostic utility of QSART
aspects of neuropathic pain. For example, patients was compared to QST and IENFD. QSART had
with pressing pain and evoked pain who did not higher sensitivity and specificity (82%, 89%)
respond to either agent did so when treated with than QST (88%, 50%) and IENFD (67%, 91%).
a combination of the two [101]. Among 29 QSART and IENFD had positive predictive val-
patients with painful DPN treated with intrave- ues of 90%, but the negative predictive value of
nous lidocaine, those with the irritable nociceptor QSART was 80%, while it was 68% for
phenotype were more likely to respond (odds IENFD.  These results suggest that both IENFD
ratio 8.67, 95% CI 1.4–54.8). These data suggest and QSART both have diagnostic value [103]. A
careful sensory phenotyping using clinical mea- smaller study focusing on DPN, and which
sures and QST may have particular utility in neu- included patients with both large and small fiber
ropathic pain clinical trials in order to establish predominant neuropathies, found IENFD to be
predictive models for therapeutic response. superior to QSART in patients with large fiber
involvement, whereas the diagnostic yield of
both was similar in those with small fiber neu-
5 Sudomotor Testing ropathy [104]. Limitations of QSART include the
requirement for specialized equipment and the
Sweat glands are innervated by postganglionic need for meticulous attention to testing parame-
cholinergic unmyelinated small fibers, thus ters, including medication use and room tempera-
explaining why peripheral neuropathies involv- ture. For these reasons, test-retest reliability of
ing small fibers also impact sudomotor function. QSART is variable, with some reports suggesting
This may be clinically evident to the patient as even marked variability [105, 106]. These factors
either reduced or increased sweating or it may be also limit the utility of QSART as an endpoint
asymptomatic. Sudomotor testing offers another measure in clinical trials. Nevertheless, in labora-
diagnostic and research tool for DPN. As is the tories with robust quality control, QSART is a
case with QST, there are a spectrum of tests rang- valuable diagnostic and research tool.
104 L. Davalos et al.

5.2 Thermoregulatory Sweat 5.4 Electrochemical Sweat


Testing Conductance (ESC)

Thermoregulatory sweat testing (TST) is per- ESC can be measured using Sudoscan™ (Impeto
formed by the application of a topical dye that Medical, Paris France), which is a novel technol-
changes color when wet (e.g., cornstarch mixed ogy that relies upon the concept of electrochemi-
with alizarin red). The patient is then placed in a cal conductance to assess for neuropathy [112].
chamber that is warmed with controlled humid- The palms and soles of the examinee are placed
ity. TST evaluates the sudomotor system in an on examining electrodes and a low-voltage con-
integrated fashion (i.e., not just postganglionic stant electrical current is applied. This current
fibers) across the entire body. The typical finding attracts sodium chloride from the sweat on exam-
in DPN is reduced sweating distally, although inee’s soles and palms onto the nickel in the
patients with diabetes may have a spectrum of examining electrodes. By comparing the ratio of
abnormalities, including regional and global current generated to current provided across the
defects due to focal neuropathies or generalized tested surface areas, electrochemical skin con-
autonomic neuropathy, respectively [107]. TST is ductance (ESC) is calculated by the device and
a valuable tool in the evaluation of patients with serves as a measure of DPN severity. While ESC
sweating disorders, but it is not commonly per- requires specialized equipment, the testing is
formed in the diagnosis of DPN. very quick and well tolerated. Several studies
suggest ESC has good diagnostic performance
for DPN [113] [114]. However, concerns have
5.3 Neuropad: Indicator been raised regarding variability in ESC values,
Plaster Test lack of age and gender differences (unlike all
other clinical and laboratory based neuropathy
Neuropad, sometimes referred to as the Indicator measures), and the degree to which ESC actually
Plaster Test, is a highly sensitive, practical, and measures small fiber function [115]. Given these
quick measure of sudomotor impairment in DPN concerns, the clinical value of ESC for DPN
[108]. When the plaster is applied to the plantar diagnosis remains uncertain.
surface of the foot, typically between the first and
second metatarsal heads, the presence of healthy
and normal sweat output produces a color change 6 Corneal Confocal Microscopy
of the cobalt II constituent in the plaster from blue
to pink (within 10 min). Lack of color change The cornea is the most densely innervated part of
within a set time threshold identifies an impairment the human body, containing myelinated Aδ and
in sudomotor function, and by extension, suggests unmyelinated C-fibers, and derives its innerva-
peripheral neuropathy. Neuropad is sensitive (65– tion from the ophthalmic division of the trigemi-
100%) in the diagnosis of DPN and has a high nal nerve. Corneal confocal microscopy (CCM)
negative predictive value (63–100%), but has poor is a noninvasive technique that allows sequential
specificity (32–78.5%) and positive predictive in  vivo observation of the small unmyelinated
value (23.3–93.2%) [109]. Meta-analytic data sug- nerve fiber bundle of the cornea, comparable to
gests an average sensitivity of 86% and specificity that obtained with histopathological examination
of 65%, while a multicenter study shows an even [116]. Images are acquired with a confocal
higher sensitivity of 94.9% and negative predictive microscope (Heidelberg HRT III with a Rostock
value of 98.1% [110]. Thus, it is a valuable and Cornea Module, Heidelberg Engineering,
convenient means of excluding DPN, but not use- Heidelberg, Germany). Typically, 5–8 high-­
ful in confirming diagnosis. The combination of quality, non-overlapping images from the central
monofilament testing with Neuropad is a cost- and peripheral-central cornea are chosen, after
effective screening strategy for DPN [111]. which nerve morphology is analyzed to deter-
Diagnostic Techniques for Diabetic Peripheral Neuropathy 105

mine nerve fiber density (NFD fibers/mm2) and tion of T1D and T2D patients, and only a few
nerve fiber length (mm/mm2) (Fig.  5) [117]. have adjusted for age [74, 129, 130], which is a
Normative data are available, which demonstrate confounder [118]. A large cross sectional study
a decline in NFD and NFL with age [118, 119]. of T2D patients from the ADDITION trial found
Many studies have shown good interobserver that CCM could not distinguish patients with
and intraobserver reliability with most parame- DPN from patients without DPN, but NFD was
ters when a single image is pre-selected [120– lower in T2D patients as compared to controls
122]. However, when trying to replicate the [131]. These results suggested that CCM might
whole process with multiple images, NFL has the have a better neuropathy diagnostic value in
best reproducibility [122]. This reinforces the patients with DM1 than in DM2. In order to
need of a standardized protocol for imaging assess the validity of CCM as a diagnostic tool
selection. Given the variability risk of manually for DPN in a larger population, a multinational
selecting corneal nerve fibers, an automated mea- cohort of 998 diabetic patients (516 with T1DM
surement software has been created. Some stud- and 482 with T2DM) was studied. The results
ies report that the automated measurement showed that NFL, NFD and NBD were signifi-
underestimates the manual measurement [123], cantly lower in T1D patients with DPN as com-
while others report a good correlation with both pared to T1D patients without DPN, and only
methods [124]. NFL and NBD were significantly lower in T2D
Numerous studies suggest CCM is a sensitive patients with DPN as compared toT2D patients
diagnostic measure of DPN [125–127]. In 2015, without DPN [124]. This data supported the find-
a meta-analysis that evaluated the utility of CCM ings of prior small cohorts, suggesting that CCM
in the assessment of DPN concluded that NFD, is a reliable diagnostic tool in both diabetic neu-
NBD and NFL are all significantly reduced in ropathy types.
patients with DPN when compared to healthy The appeal of CCM as a diagnostic and
controls and diabetic patients without DPN [128]. research tool is that it assesses the same fiber
However, most positive studies have traditionally class as IENFD in a noninvasive and easily
focused on small cohorts of T1D or a combina- repeatable fashion. Studies exploring the correla-

a b

Fig. 5  Corneal confocal microscopy (CCM) from a nor- demonstrates reduced nerve fiber length and density as
mal subject demonstrates normal nerve fiber length and well as proliferation of Langerhan’s Cells (arrowheads)
density (a). CCM from a patient with diabetic neuropathy (b)
106 L. Davalos et al.

tion between CCM and IENFD provide a valu- sary to determine its clinical meaning and role as
able window into the biological meaning of an outcome measure in clinical trials, it remains a
CCM, particularly since DPN clinically involves promising research tool.
the longest nerves in the body, while CCM mea-
sures some of the shortest nerves. Several small
studies suggest CCM performs comparably well 7 Conclusion
to IEFND in patients with T1D [74, 129], while
another study did not find a correlation between The clinical diagnosis of DPN is based on a com-
CCM and IENFD in T2D patients [132]. A third bination of symptoms and signs and can be sup-
study found that CCM’s diagnostic performance ported by application of several diagnostic tests
was slightly lower than IENFD for DPN [133]. that assess both small and large fiber function
While larger longitudinal studies comparing (Table 2). While these tests are not always neces-
CCM to other neuropathy measures are neces- sary in settings where patients present with typi-

Table 2  Diagnostic techniques for diabetic peripheral neuropathy


Diagnostic Nerve fiber type
technique target Advantages Disadvantages
Nerve conduction Large diameter Objective, high intra-observer Do not assess small fibers, high
studies myelinated reliability for conduction interobserver variability, require referral
fibers velocity measurement, widely to a neurophysiology lab with trained
available, gold standard for examiners
large fiber neuropathy
Skin biopsy Small diameter Objective, gold standard for Minimally invasive, technically
(IEFND) unmyelinated small fiber neuropathy, demanding requiring an expert
nerve fibers strongest research outcome reference laboratory with robust quality
measure for DPN, high control procedures
intraobserver and interobserver
reliability, can assess early
reinnervation
Quantitative Small and large Fast and easy to perform, Subjective, costly, requires specialized
sensory testing nerve fibers consistent inter and intra rater equipment, abnormality does not
reliability, useful in necessarily localize to the peripheral
phenotyping pain and nervous system
predicting treatment response.
The only technique that
captures the patient experience
Sudomotor Autonomic Fast and easy to perform, Test-retest reliability can vary due to
testing (QSART) small diameter objective, reproducible external factors (e.g. medications), role
unmyelinated limited in research, requires specialized
fibers equipment
Electrochemical Autonomic Fast and easy to perform Requires specialized equipment. High
Sweat small diameter degree of test retest variability, lack of
Conductance unmyelinated age and gender differences (unlike all
fibers other clinical and laboratory based
neuropathy measures), and the degree to
which it actually measures small fiber
function
Corneal confocal Small diameter Noninvasive, reproducible, Not widely available, need for further
microscopy unmyelinated objective, and easily repeatable validation, technique assesses short
nerve fibers length nerve fibers in a disease that is
generally viewed as length dependent
(i.e., involving the longest fibers)
IEFND intraepidermal nerve fiber density, QSART quantitative sudomotor axon reflex testing, PNS peripheral nervous
system
Diagnostic Techniques for Diabetic Peripheral Neuropathy 107

cal clinical features of DPN, they can be very measures have particular value as research tools.
useful in situations where there are atypical clini- While NCS remain the mainstay for the clinical
cal features. Some are also well suited at screen- evaluation of large fiber neuropathy and IENFD
ing patients for early or preclinical neuropathy or is the current gold standard for small fiber neu-
to identify those at risk for important health out- ropathy, a variety of point of care devices and
comes (e.g., foot ulceration), and variety of point tools and newer promising techniques, including
of care devices are available (Table  3). Many CCM, are now available.

Table 3  Point of care devices for diagnosing diabetic peripheral neuropathy


Diagnostic Nerve fiber type
technique Device target Advantages Disadvantages
Nerve Point of care Large nerve High sensitivity, Overestimates sural nerve
conduction nerve fibers acceptable specificity, conduction velocity
studies conduction good intra and inter rater
studies reliability, fast,
inexpensive, easy to use
Quantitative Vibrometer Large nerve Portable, inexpensive,
sensory fibers easy to perform,
testing associates with
neuropathy disability
score
NeuroQuick Small Portable, discriminates Low sensitivity for painful SFN
myelinated between DPN and compared with standard QST
nerve fibers control subjects, more cold detection threshold; not
(cold detection) sensitive than thermal adequate as a stand-alone
perception threshold measure of impairment in cold
perception
NerveCheck Large and small Portable, good test-retest
nerve fibers reliability, comparable
diagnostic accuracy to
traditional QST
Neurometer Large Discriminates DPN from Reproducibility better in controls
myelinated and control subjects well than diabetic patients. Sensitivity
small may be low
myelinated
nerve fibers
Sudomotor Neuropad Autonomic Sensitive and high Low specificity, not useful at
function small nerve negative predictive value, confirming diagnosis of DPN
fibers fast, easy to perform, and
well tolerated
108 L. Davalos et al.

References 15. Amthor KF, Dahl-Jørgensen K, Berg TJ, Heier MS,


Sandvik L, Aagenaes O, et  al. The effect of 8 years
of strict glycaemic control on peripheral nerve func-
1. Tesfaye S, Boulton AJM, Dyck PJ, Freeman R,
tion in IDDM patients: the Oslo Study. Diabetologia.
Horowitz M, Kempler P, et  al. Diabetic neuropa-
1994;37(6):579–84.
thies: update on definitions, diagnostic criteria, esti-
16. Andersen H, Stålberg E, Falck B.  F-wave latency,
mation of severity, and treatments. Diabetes Care.
the most sensitive nerve conduction parameter
2010;33(10):2285–93.
in patients with diabetes mellitus. Muscle Nerve.
2. Smith AG. Do all neuropathy patients need an EMG at
1997;20(10):1296–302.
least once? Contin Minneap Minn. 2014;20(5):1430–4.
17. Kohara N, Kimura J, Kaji R, Goto Y, Ishii J, Takiguchi
3. Divisova S, Vlckova E, Hnojcikova M, Skorna
M, et  al. F-wave latency serves as the most repro-
M, Nemec M, Dubovy P, et  al. Prediabetes/early
ducible measure in nerve conduction studies of dia-
diabetes-associated neuropathy predominantly
betic polyneuropathy: multicentre analysis in healthy
involves sensory small fibres. J Peripher Nerv Syst.
subjects and patients with diabetic polyneuropathy.
2012;17(3):341–50.
Diabetologia. 2000;43(7):915–21.
4. Singleton JR, Bixby B, Russell JW, Feldman EL,
18. Dyck PJ, Lais A, Karnes JL, O’Brien P, Rizza R. Fiber
Peltier A, Goldstein J, et  al. The Utah early neu-
loss is primary and multifocal in sural nerves in diabetic
ropathy scale: a sensitive clinical scale for early sen-
polyneuropathy. Ann Neurol. 1986;19(5):425–39.
sory predominant neuropathy. J Peripher Nerv Syst.
19. Behse F, Buchthal F, Carlsen F.  Nerve biopsy and
2008;13(3):218–27.
conduction studies in diabetic neuropathy. J Neurol
5. Andersson C, Guttorp P, Särkkä A. Discovering early
Neurosurg Psychiatry. 1977;40(11):1072–82.
diabetic neuropathy from epidermal nerve fiber pat-
20. Misawa S, Kuwabara S, Kanai K, Tamura N, Hiraga
terns. Stat Med. 2016;35(24):4427–42.
A, Nakata M, et  al. Axonal potassium conductance
6. Devigili G, Tugnoli V, Penza P, Camozzi F, Lombardi
and glycemic control in human diabetic nerves.
R, Melli G, et al. The diagnostic criteria for small fibre
Clin Neurophysiol Off J Int Fed Clin Neurophysiol.
neuropathy: from symptoms to neuropathology. Brain
2005;116(5):1181–7.
J Neurol. 2008;131(7):1912–25.
21. Yagihashi S, Yamagishi S-I, Wada R.  Pathology and
7. Lauria G, Lombardi R, Camozzi F, Devigili G. Skin
pathogenetic mechanisms of diabetic neuropathy: cor-
biopsy for the diagnosis of peripheral neuropathy.
relation with clinical signs and symptoms. Diabetes
Histopathology. 2009;54(3):273–85.
Res Clin Pract. 2007;77(1):184–9.
8. Sivaskandarajah GA, Halpern EM, Lovblom LE,
22. Tomlinson DR.  Mitogen-activated protein kinases
Weisman A, Orlov S, Bril V, et al. Structure-function
as glucose transducers for diabetic complications.
relationship between corneal nerves and conventional
Diabetologia. 1999;42(11):1271–81.
small-fiber tests in type 1 diabetes. Diabetes Care.
23. American Diabetes Association American Academy
2013;36(9):2748–55.
of Neurology. Consensus statement: report and rec-
9. Callaghan BC, Price RS, Chen KS, Feldman EL. The
ommendations of the San Antonio conference on dia-
importance of rare subtypes in diagnosis and treat-
betic neuropathy. Diabetes Care. 1988;11(7):592–7.
ment of peripheral neuropathy: a review. JAMA
24. England JD, Gronseth GS, Franklin G, Miller RG,
Neurol. 2015;72(12):1510–8.
Asbury AK, Carter GT, et al. Distal symmetric poly-
10. Gooch CL, Weimer LH. The electrodiagnosis of neu-
neuropathy: a definition for clinical research: report of
ropathy: basic principles and common pitfalls. Neurol
the American Academy of Neurology, the American
Clin. 2007;25(1):1–28.
Association of Electrodiagnostic Medicine, and
11. Weisman A, Bril V, Ngo M, Lovblom LE, Halpern
the American Academy of Physical Medicine and
EM, Orszag A, et  al. Identification and prediction
Rehabilitation. Neurology. 2005;64(2):199–207.
of diabetic sensorimotor polyneuropathy using indi-
25. Dyck PJ, Carter RE, Litchy WJ. Modeling nerve con-
vidual and simple combinations of nerve conduction
duction criteria for diagnosis of diabetic polyneuropa-
study parameters. PLoS One. 2013;8(3):e58783.
thy. Muscle Nerve. 2011;44(3):340–5.
12. Russell JW, Karnes JL, Dyck PJ. Sural nerve myelin-
26. Callaghan B, McCammon R, Kerber K, Xu X, Langa
ated fiber density differences associated with mean-
KM, Feldman E. Tests and expenditures in the initial
ingful changes in clinical and electrophysiologic
evaluation of peripheral neuropathy. Arch Intern Med.
measurements. J Neurol Sci. 1996;135(2):114–7.
2012;172(2):127–32.
13. Partanen J, Niskanen L, Lehtinen J, Mervaala E,
27. Callaghan BC, Kerber KA, Lisabeth LL, Morgenstern
Siitonen O, Uusitupa M. Natural history of peripheral
LB, Longoria R, Rodgers A, et  al. Role of neu-
neuropathy in patients with non-­ insulin-dependent
rologists and diagnostic tests on the management of
diabetes mellitus. N Engl J Med. 1995;333(2):89–94.
distal symmetric polyneuropathy. JAMA Neurol.
14. Nathan DM, Genuth S, Lachin J, Cleary P, Crofford
2014;71(9):1143–9.
O, et  al. The effect of intensive treatment of diabe-
28. Pop-Busui R, Boulton AJM, Feldman EL, Bril V,
tes on the development and progression of long-term
Freeman R, Malik RA, et  al. Diabetic neuropa-
complications in insulin-dependent diabetes mellitus.
thy: a position statement by the American Diabetes
N Engl J Med. 1993;329(14):977–86.
Association. Diabetes Care. 2017;40(1):136–54.
Diagnostic Techniques for Diabetic Peripheral Neuropathy 109

29. Mulder DW, Lambert EH, Bastron JA, Sprague 44. Boulton AJ, Knight G, Drury J, Ward JD.  The
RG.  The neuropathies associated with diabetes prevalence of symptomatic, diabetic neuropathy
mellitus. A clinical and electromyographic study in an insulin-treated population. Diabetes Care.
of 103 unselected diabetic patients. Neurology. 1985;8(2):125–8.
1961;11(4):275–84. 45. Dyck PJ, Litchy WJ, Daube JR, Harper CM, Dyck
30. Lamontagne A, Buchthal F.  Electrophysiological PJB, Davies J, et  al. Individual attributes versus
studies in diabetic neuropathy. J Neurol Neurosurg composite scores of nerve conduction abnormality:
Psychiatry. 1970;33(4):442–52. sensitivity, reproducibility, and concordance with
31. Dyck PJ, Davies JL, Litchy WJ, O’Brien impairment. Muscle Nerve. 2003;27(2):202–10.
PC.  Longitudinal assessment of diabetic polyneu- 46. Feldman EL, Stevens MJ, Thomas PK, Brown MB,
ropathy using a composite score in the Rochester Canal N, Greene DA.  A practical two-step quantita-
Diabetic Neuropathy Study cohort. Neurology. tive clinical and electrophysiological assessment for
1997;49(1):229–39. the diagnosis and staging of diabetic neuropathy.
32. Dyck PJ, Karnes JL, O’Brien PC, Litchy WJ, Low Diabetes Care. 1994;17(11):1281–9.
PA, Melton LJ.  The Rochester Diabetic Neuropathy 47. Dyck PJ, Albers JW, Andersen H, Arezzo JC,
Study: reassessment of tests and criteria for diagnosis Biessels G-J, Bril V, et al. Diabetic polyneuropathies:
and staged severity. Neurology. 1992;42(6):1164–70. update on research definition, diagnostic criteria
33. Author. Effect of intensive diabetes treatment on nerve and estimation of severity. Diabetes Metab Res Rev.
conduction in the diabetes control and complications 2011;27(7):620–8.
trial. Ann Neurol. 1995;38(6):869–80. 48. Malik RA, Williamson S, Abbott C, Carrington
34. Bril V.  Role of electrophysiological studies in dia- AL, Iqbal J, Schady W, et  al. Effect of angiotensin-­
betic neuropathy. Can J Neurol Sci J Can Sci Neurol. converting-­enzyme (ACE) inhibitor trandolapril on
1994;21(4):8–12. human diabetic neuropathy: randomised double-blind
35. Bril V, Ellison R, Ngo M, Bergstrom B, Raynard D, controlled trial. Lancet. 1998;352(9145):1978–81.
Gin H. Electrophysiological monitoring in clinical tri- 49. Wahren J, Foyt H, Daniels M, Arezzo JC.  Long-­
als. Roche Neuropathy Study Group. Muscle Nerve. acting c-peptide and neuropathy in type 1 dia-
1998;21(11):1368–73. betes: a 12-month clinical trial. Diabetes Care.
36. Dyck PJ, Overland CJ, Low PA, Litchy WJ, Davies 2016;39(4):596–602.
JL, Dyck PJB, et  al. Signs and symptoms versus 50. Ruggenenti P, Lauria G, Iliev IP, Fassi A, Ilieva AP,
nerve conduction studies to diagnose diabetic sen- Rota S, et  al. Effects of manidipine and delapril in
sorimotor polyneuropathy: Cl vs. Muscle Nerve. hypertensive patients with type 2 diabetes mellitus:
2010;42(2):157–64. the delapril and manidipine for nephroprotection
37. Perkins BA, Greene DA, Bril V.  Glycemic con- in diabetes (DEMAND) randomized clinical trial.
trol is related to the morphological severity of dia- Hypertens Dallas Tex. 2011;58(5):776–83.
betic sensorimotor polyneuropathy. Diabetes Care. 51. Dyck PJ, Norell JE, Tritschler H, Schuette K,
2001;24(4):748–52. Samigullin R, Ziegler D, et  al. Challenges in design
38. Perkins BA, Olaleye D, Zinman B, Bril V.  Simple of multicenter trials: end points assessed longitudi-
screening tests for peripheral neuropathy in the diabe- nally for change and monotonicity. Diabetes Care.
tes clinic. Diabetes Care. 2001;24(2):250–6. 2007;30(10):2619–25.
39. Perkins BA, Bril V.  Diabetic neuropathy: a review 52. Polydefkis M, Hauer P, Sheth S, Sirdofsky M, Griffin
emphasizing diagnostic methods. Clin Neurophysiol JW, McArthur JC. The time course of epidermal nerve
Off J Int Fed Clin Neurophysiol. 2003;114(7):1167–75. fibre regeneration: studies in normal controls and in
40. Dyck PJ, Litchy WJ, Lehman KA, Hokanson JL, Low people with diabetes, with and without neuropathy.
PA, O’Brien PC.  Variables influencing neuropathic Brain J Neurol. 2004;127(7):1606–15.
endpoints: the Rochester Diabetic Neuropathy Study 53. Lee JA, Halpern EM, Lovblom LE, Yeung E, Bril V,
of Healthy Subjects. Neurology. 1995;45(6):1115–21. Perkins BA. Reliability and validity of a point-of-care
41. Dyck PJ, Albers JW, Wolfe J, Bolton CF, Walsh N, sural nerve conduction device for identification of dia-
Klein CJ, et  al. A trial of proficiency of nerve con- betic neuropathy. PLoS One. 2014;9(1):e86515.
duction: greater standardization still needed. Muscle 54. Langerhans P.  Ueber die Nerven der menschlichen
Nerve. 2013;48(3):369–74. Haut. Arch Für Pathol Anat Physiol Für Klin Med.
42. Litchy WJ, Albers JW, Wolfe J, Bolton CF, Walsh N, 1868;44(2):325–37.
Klein CJ, et al. Proficiency of nerve conduction using 55. Dyck PJ, Winkelmann RK, Bolton CF. Quantitation
standard methods and reference values (Cl. NPhys of Meissner’s corpuscles in hereditary neurologic
Trial 4). Muscle Nerve. 2014;50(6):900–8. disorders. Charcot-Marie-Tooth disease, Roussy-­
43. Dyck PJ, Kratz KM, Karnes JL, Litchy WJ, Klein Levy syndrome, Dejerine-Sottas disease, hereditary
R, Pach JM, et al. The prevalence by staged severity sensory neuropathy, spinocerebellar degenerations,
of various types of diabetic neuropathy, retinopathy, and hereditary spastic paraplegia. Neurology.
and nephropathy in a population-based cohort: the 1966;16(1):10–7.
Rochester Diabetic Neuropathy Study. Neurology. 56. Dalsgaard CJ, Rydh M, Haegerstrand A.  Cutaneous
1993;43(4):817–24. innervation in man visualized with protein gene
110 L. Davalos et al.

product 9.5 (PGP 9.5) antibodies. Histochemistry. 71. Sorensen L, Molyneaux L, Yue DK. The relationship
1989;92(5):385–90. among pain, sensory loss, and small nerve fibers in
57. Kanitakis J.  Anatomy, histology and immunohisto- diabetes. Diabetes Care. 2006;29(4):883–7.
chemistry of normal human skin. Eur J Dermatol. 72. Nebuchennykh M, Løseth S, Lindal S, Mellgren
2002;12(4):390–9; quiz 400–1 SI. The value of skin biopsy with recording of intraepi-
58. Wendelschafer-Crabb G, Kennedy WR, Walk dermal nerve fiber density and quantitative sensory
D. Morphological features of nerves in skin biopsies. testing in the assessment of small fiber involvement
J Neurol Sci. 2006;242(1–2):15–21. in patients with different causes of polyneuropathy. J
59. McArthur JC, Stocks EA, Hauer P, Cornblath DR, Neurol. 2009;256(7):1067–75.
Griffin JW.  Epidermal nerve fiber density: norma- 73. Vlcková-Moravcová E, Bednarík J, Dusek L, Toyka
tive reference range and diagnostic efficiency. Arch KV, Sommer C.  Diagnostic validity of epidermal
Neurol. 1998;55(12):1513–20. nerve fiber densities in painful sensory neuropathies.
60. Liu Y, Fan X, Wei Y, Piao Z, Jiang X. Intraepidermal Muscle Nerve. 2008;37(1):50–60.
nerve fiber density of healthy human. Neurol Res. 74. Alam U, Jeziorska M, Petropoulos IN, Asghar O,
2014;36(10):911–4. Fadavi H, Ponirakis G, et  al. Diagnostic utility of
61. Lauria G, Cornblath DR, Johansson O, McArthur JC, corneal confocal microscopy and intra-epidermal
Mellgren SI, Nolano M, et al. EFNS guidelines on the nerve fibre density in diabetic neuropathy. PLoS One.
use of skin biopsy in the diagnosis of peripheral neu- 2017;12(7):e0180175.
ropathy. Eur J Neurol. 2005;12(10):747–58. 75. Griffin JW, Thompson WJ.  Biology and pathol-
62. Lauria G, Bakkers M, Schmitz C, Lombardi R, Penza ogy of nonmyelinating Schwann cells. Glia.
P, Devigili G, et  al. Intraepidermal nerve fiber den- 2008;56(14):1518–31.
sity at the distal leg: a worldwide normative reference 76. Smith AG, Russell J, Feldman EL, Goldstein J, Peltier
study. J Peripher Nerv Syst. 2010;15(3):202–7. A, Smith S, et  al. Lifestyle intervention for pre-dia-
63. Smith AG, Howard JR, Kroll R, Ramachandran P, betic neuropathy. Diabetes Care. 2006;29(6):1294–9.
Hauer P, Singleton JR, et  al. The reliability of skin 77. Weintraub MI, Herrmann DN, Smith AG, Backonja
biopsy with measurement of intraepidermal nerve MM, Cole SP. Pulsed electromagnetic fields to reduce
fiber density. J Neurol Sci. 2005;228(1):65–9. diabetic neuropathic pain and stimulate neuronal
64. Stocks EA, McArthur JC, Griffen JW, Mouton PR. An repair: a randomized controlled trial. Arch Phys Med
unbiased method for estimation of total epidermal Rehabil. 2009;90(7):1102–9.
nerve fibre length. J Neurocytol. 1996;25(11):637–44. 78. Rajan B, Polydefkis M, Hauer P, Griffin JW, McArthur
65. Hilliges M, Johansson O.  Comparative analysis of JC.  Epidermal reinnervation after intracutaneous
numerical estimation methods of epithelial nerve axotomy in man. J Comp Neurol. 2003;457(1):24–36.
fibers using tissue sections. J Peripher Nerv Syst. 79. Simone DA, Nolano M, Johnson T, Wendelschafer-­
1999;4(1):53–7. Crabb G, Kennedy WR.  Intradermal injection of
66. Koskinen M, Hietaharju A, Kyläniemi M, Peltola J, capsaicin in humans produces degeneration and
Rantala I, Udd B, et al. A quantitative method for the subsequent reinnervation of epidermal nerve fibers:
assessment of intraepidermal nerve fibers in small-­ correlation with sensory function. J Neurosci.
fiber neuropathy. J Neurol. 2005;252(7):789–94. 1998;18(21):8947–59.
67. Joint Task Force of the EFNS and the PNS. European 80. Nolano M, Simone DA, Wendelschafer-Crabb G,
Federation of Neurological Societies/Peripheral Nerve Johnson T, Hazen E, Kennedy WR. Topical capsaicin
Society Guideline on the use of skin biopsy in the in humans: parallel loss of epidermal nerve fibers and
diagnosis of small fiber neuropathy. Report of a joint pain sensation. Pain. 1999;81(1–2):135–45.
task force of the European Federation of Neurological 81. Siao P, Cros DP.  Quantitative sensory testing. Phys
Societies and the Peripheral Nerve Society. J Peripher Med Rehabil Clin N Am. 2003;14(2):261–86.
Nerv Syst. 2010;15(2):79–92. 82. Dyck PJ, Argyros B, Russell JW, Gahnstrom LE,
68. Khoshnoodi MA, Truelove S, Burakgazi A, Hoke A, Nalepa S, Albers JW, et al. Multicenter trial of the pro-
Mammen AL, Polydefkis M.  Longitudinal assess- ficiency of smart quantitative sensation tests. Muscle
ment of small fiber neuropathy: evidence of a non-­ Nerve. 2014;49(5):645–53.
length-­dependent distal axonopathy. JAMA Neurol. 83. Krumova EK, Geber C, Westermann A, Maier
2016;73(6):684–90. C.  Neuropathic pain: is quantitative sensory testing
69. Quattrini C, Tavakoli M, Jeziorska M, Kallinikos P, helpful? Curr Diab Rep. 2012;12(4):393–402.
Tesfaye S, Finnigan J, et  al. Surrogate markers of 84. Brown MJ, Bird SJ, Watling S, Kaleta H, Hayes L,
small fiber damage in human diabetic neuropathy. Eckert S, et al. Natural progression of diabetic periph-
Diabetes. 2007;56(8):2148–54. eral neuropathy in the Zenarestat study population.
70. Løseth S, Mellgren SI, Jorde R, Lindal S, Stålberg Diabetes Care. 2004;27(5):1153–9.
E.  Polyneuropathy in type 1 and type 2 diabetes: 85. Periquet MI, Novak V, Collins MP, Nagaraja HN,
comparison of nerve conduction studies, thermal per- Erdem S, Nash SM, et al. Painful sensory neuropathy:
ception thresholds and intraepidermal nerve fibre den- prospective evaluation using skin biopsy. Neurology.
sities. Diabetes Metab Res Rev. 2010;26(2):100–6. 1999;53(8):1641–7.
Diagnostic Techniques for Diabetic Peripheral Neuropathy 111

86. Fagius J, Wahren LK.  Variability of sensory thresh- 99. Maier C, Baron R, Tölle TR, Binder A, Birbaumer
old determination in clinical use. J Neurol Sci. N, Birklein F, et  al. Quantitative sensory testing
1981;51(1):11–27. in the German Research Network on Neuropathic
87. Shy ME, Frohman EM, So YT, Arezzo JC, Cornblath Pain (DFNS): somatosensory abnormalities in 1236
DR, Giuliani MJ, et  al. Quantitative sensory test- patients with different neuropathic pain syndromes.
ing: report of the Therapeutics and Technology Pain. 2010;150(3):439–50.
Assessment Subcommittee of the American Academy 100. Themistocleous AC, Ramirez JD, Shillo PR, Lees
of Neurology. Neurology. 2003;60(6):898–904. JG, Selvarajah D, Orengo C, et  al. The Pain in
88. Bowling FL, Abbott CA, Harris WE, Atanasov S, Neuropathy Study (PiNS): a cross-sectional obser-
Malik RA, Boulton AJM. A pocket-sized disposable vational study determining the somatosensory phe-
device for testing the integrity of sensation in the notype of painful and painless diabetic neuropathy.
outpatient setting. Diabet Med J Br Diabet Assoc. Pain. 2016;157(5):1132–45.
2012;29(12):1550–2. 101. Bouhassira D, Wilhelm S, Schacht A, Perrot S,
89. Bracewell N, Game F, Jeffcoate W, Scammell Kosek E, Cruccu G, et  al. Neuropathic pain phe-
BE. Clinical evaluation of a new device in the assess- notyping as a predictor of treatment response in
ment of peripheral sensory neuropathy in diabetes. painful diabetic neuropathy: data from the ran-
Diabet Med J Br Diabet Assoc. 2012;29(12):1553–5. domized, double-blind, COMBO-DN study. Pain.
90. Ziegler D, Siekierka-Kleiser E, Meyer B, Schweers 2014;155(10):2171–9.
M.  Validation of a novel screening device 102. Low VA, Sandroni P, Fealey RD, Low PA. Detection
(NeuroQuick) for quantitative assessment of small of small-fiber neuropathy by sudomotor testing.
nerve fiber dysfunction as an early feature of diabetic Muscle Nerve. 2006;34(1):57–61.
polyneuropathy. Diabetes Care. 2005;28(5):1169–74. 103. Thaisetthawatkul P, Fernandes Filho JAM,
91. Haussleiter IS, Richter H, Scherens A, Schwenkreis P, Herrmann DN.  Contribution of QSART to the
Tegenthoff M, Maier C. NeuroQuick–a novel bedside diagnosis of small fiber neuropathy. Muscle Nerve.
test for small fiber neuropathy? Eur J Pain Lond Engl. 2013;48(6):883–8.
2008;12(8):1000–7. 104. Abuzinadah AR, Kluding P, Wright D, D’Silva L,
92. Ponirakis G, Odriozola MN, Odriozola S, Petropoulos Ryals J, Hendry B, et  al. Less is more in diabetic
IN, Azmi S, Fadavi H, et al. NerveCheck: an inexpen- neuropathy diagnosis: comparison of quantita-
sive quantitative sensory testing device for patients tive sudomotor axon reflex and skin biopsy. J Clin
with diabetic neuropathy. Diabetes Res Clin Pract. Neuromuscul Dis. 2017;19(1):5–11.
2016;113:101–7. 105. Berger MJ, Kimpinski K.  Test-retest reliability of
93. Inceu GV, Veresiu IA.  Measurement of current quantitative sudomotor axon reflex testing. J Clin
perception thresholds using the Neurometer®  - Neurophysiol Off Publ Am Electroencephalogr Soc.
applicability in diabetic neuropathy. Clujul Med. 2013;30(3):308–12.
2015;88(4):449–52. 106. Peltier A, Smith AG, Russell JW, Sheikh K, Bixby B,
94. Lv S, Fang C, Hu J, Huang Y, Yang B, Zou R, et al. Howard J, et al. Reliability of quantitative sudomo-
Assessment of peripheral neuropathy using measure- tor axon reflex testing and quantitative sensory test-
ment of the current perception threshold with the ing in neuropathy of impaired glucose regulation.
Neurometer® in patients with type 1 diabetes melli- Muscle Nerve. 2009;39(4):529–35.
tus. Diabetes Res Clin Pract. 2015;109(1):130–4. 107. Fealey RD, Low PA, Thomas JE. Thermoregulatory
95. Matsutomo R, Takebayashi K, Aso Y.  Assessment sweating abnormalities in diabetes mellitus. Mayo
of peripheral neuropathy using measurement of the Clin Proc. 1989;64(6):617–28.
current perception threshold with the neurometer in 108. Papanas N, Papatheodorou K, Christakidis D,
patients with type 2 diabetes mellitus. J Int Med Res. Papazoglou D, Giassakis G, Piperidou H, et  al.
2005;33(4):442–53. Evaluation of a new indicator test for sudomotor
96. Pitei DL, Watkins PJ, Stevens MJ, Edmonds ME. The function (Neuropad) in the diagnosis of peripheral
value of the Neurometer in assessing diabetic neurop- neuropathy in type 2 diabetic patients. Exp Clin
athy by measurement of the current perception thresh- Endocrinol Diabetes Off J Ger Soc Endocrinol Ger
old. Diabet Med J Br Diabet Assoc. 1994;11(9):872–6. Diabetes Assoc. 2005;113(4):195–8.
97. Koo BK, Ohn JH, Kwak S-H, Moon MK. Assessment 109. Papanas N, Boulton AJM, Malik RA, Manes
of diabetic polyneuropathy and autonomic neuropa- C, Schnell O, Spallone V, et  al. A simple new
thy using current perception threshold in Korean ­non-­invasive sweat indicator test for the diagnosis of
patients with diabetes mellitus. Diabetes Metab J. diabetic neuropathy. Diabet Med J Br Diabet Assoc.
2014;38(4):285–93. 2013;30(5):525–34.
98. Gierthmühlen J, Binder A, Förster M, Baron R.  Do 110. Tsapas A, Liakos A, Paschos P, Karagiannis T,
we measure what patients feel? An analysis of cor- Bekiari E, Tentolouris N, et  al. A simple plaster
respondence between somatosensory modalities upon for screening for diabetic neuropathy: a diagnostic
quantitative sensory testing and self-reported pain test accuracy systematic review and meta-analysis.
experience. Clin J Pain. 2018;34(7):610–7. Metabolism. 2014;63(4):584–92.
112 L. Davalos et al.

111. Rodríguez-Sánchez B, Peña-Longobardo 123. Scarr D, Lovblom LE, Ostrovski I, Kelly D, Wu T,


LM, Sinclair AJ.  Cost-effectiveness analy- Farooqi MA, et  al. Agreement between automated
sis of the Neuropad device as a screening tool and manual quantification of corneal nerve fiber
for early diabetic peripheral neuropathy. Eur J length: implications for diabetic neuropathy research.
Health Econ HEPAC Health Econ Prev Care. J Diabetes Complicat. 2017;31(6):1066–73.
2020;21(3):335–49. 124. Perkins BA, Lovblom LE, Bril V, Scarr D, Ostrovski
112. Calvet JH, Dupin J, Winiecki H, Schwarz I, Orszag A, et al. Corneal confocal microscopy for
PEH. Assessment of small fiber neuropathy through identification of diabetic sensorimotor polyneu-
a quick, simple and non invasive method in a German ropathy: a pooled multinational consortium study.
diabetes outpatient clinic. Exp Clin Endocrinol Diabetologia. 2018;61(8):1856–61.
Diabetes Off J Ger Soc Endocrinol Ger Diabetes 125. Ahmed A, Bril V, Orszag A, Paulson J, Yeung E,
Assoc. 2013;121(2):80–3. Ngo M, et  al. Detection of diabetic sensorimotor
113. Casellini CM, Parson HK, Richardson MS, Nevoret polyneuropathy by corneal confocal microscopy in
ML, Vinik AI.  Sudoscan, a noninvasive tool for type 1 diabetes: a concurrent validity study. Diabetes
detecting diabetic small fiber neuropathy and Care. 2012;35(4):821–8.
autonomic dysfunction. Diabetes Technol Ther. 126. Tavakoli M, Quattrini C, Abbott C, Kallinikos P,
2013;15(11):948–53. Marshall A, Finnigan J, et  al. Corneal confocal
114. Krieger S-M, Reimann M, Haase R, Henkel E, microscopy: a novel noninvasive test to diagnose and
Hanefeld M, Ziemssen T. Sudomotor testing of dia- stratify the severity of human diabetic neuropathy.
betes polyneuropathy. Front Neurol. 2018;9:803. Diabetes Care. 2010;33(8):1792–7.
115. Rajan S, Campagnolo M, Callaghan B, Gibbons 127. Keller JM, Noben-Trauth K. Genome-wide linkage
CH. Sudomotor function testing by electrochemical analyses identify Hfhl1 and Hfhl3 with frequency-­
skin conductance: does it really measure sudomotor specific effects on the hearing spectrum of NIH
function? Clin Auton Res Off J Clin Auton Res Soc. Swiss mice. BMC Genet. 2012;13:32.
2019;29(1):31–9. 128. Jiang M-S, Yuan Y, Gu Z-X, Zhuang S-L.  Corneal
116. Oliveira-Soto L, Efron N.  Morphology of cor- confocal microscopy for assessment of diabetic
neal nerves using confocal microscopy. Cornea. peripheral neuropathy: a meta-analysis. Br J
2001;20(4):374–84. Ophthalmol. 2016 Jan;100(1):9–14.
117. Petropoulos IN, Alam U, Fadavi H, Marshall A, 129. Chen X, Graham J, Dabbah MA, Petropoulos IN,
Asghar O, Dabbah MA, et al. Rapid automated diag- Ponirakis G, Asghar O, et al. Small nerve fiber quan-
nosis of diabetic peripheral neuropathy with in vivo tification in the diagnosis of diabetic sensorimo-
corneal confocal microscopy. Invest Ophthalmol Vis tor polyneuropathy: comparing corneal confocal
Sci. 2014;55(4):2071–8. microscopy with intraepidermal nerve fiber density.
118. Tavakoli M, Ferdousi M, Petropoulos IN, Morris J, Diabetes Care. 2015;38(6):1138–44.
Pritchard N, Zhivov A, et  al. Normative values for 130. Stem MS, Hussain M, Lentz SI, Raval N, Gardner
corneal nerve morphology assessed using corneal TW, Pop-Busui R, et  al. Differential reduction in
confocal microscopy: a multinational normative data corneal nerve fiber length in patients with type 1
set. Diabetes Care. 2015;38(5):838–43. or type 2 diabetes mellitus. J Diabetes Complicat.
119. Dehghani C, Pritchard N, Edwards K, Vagenas D, 2014;28(5):658–61.
Russell AW, Malik RA, et al. Morphometric stability 131. Andersen ST, Grosen K, Tankisi H, Charles M,
of the corneal subbasal nerve plexus in healthy indi- Andersen NT, Andersen H, et  al. Corneal confocal
viduals: a 3-year longitudinal study using corneal microscopy as a tool for detecting diabetic polyneu-
confocal microscopy. Invest Ophthalmol Vis Sci. ropathy in a cohort with screen-detected type 2 dia-
2014;55(5):3195–9. betes: ADDITION-Denmark. J Diabetes Complicat.
120. Petropoulos IN, Manzoor T, Morgan P, Fadavi H, 2018;32(12):1153–9.
Asghar O, Alam U, et  al. Repeatability of in  vivo 132. Ziegler D, Papanas N, Zhivov A, Allgeier S, Winter
corneal confocal microscopy to quantify corneal K, Ziegler I, et al. Early detection of nerve fiber loss
nerve morphology. Cornea. 2013;32(5):83–9. by corneal confocal microscopy and skin biopsy
121. Efron N, Edwards K, Roper N, Pritchard N, in recently diagnosed type 2 diabetes. Diabetes.
Sampson GP, Shahidi AM, et  al. Repeatability of 2014;63(7):2454–63.
measuring corneal subbasal nerve fiber length in 133. Smith AG, Kowalsky G, Hauer P, Aperghis A,
individuals with type 2 diabetes. Eye Contact Lens. Singleton J. The diagnostic performance and clinical
2010;36(5):245–8. relevance of corneal confocal microscopy (CCM)
122. Hertz P, Bril V, Orszag A, Ahmed A, Ng E, Nwe P, in patients with diabetic peripheral neuropathy
et  al. Reproducibility of in  vivo corneal confocal (S44.001). Neurology. 2016;86:16. Available from
microscopy as a novel screening test for early dia- https://n.neurology.org/content/86/16_Supplement/
betic sensorimotor polyneuropathy. Diabet Med J Br S44.001
Diabet Assoc. 2011;28(10):1253–60.
Sensory Profiles and Diabetic
Neuropathy

Juliane Sachau, Manon Sendel, and Ralf Baron

1 Introduction a particular pain therapy and how can these


responders be identified a priori?
Up to 50% of patients with diabetic neuropathy The correct classification of neuropathic pain
suffer from neuropathic pain [1], i.e., pain that is a prerequisite to answer this question. The fact
arises after a lesion or disease of the somatosen- that different pathomechanisms can be present
sory nervous system. Neuropathic pain can pro- within one pain etiology, while the very same
voke sleep disturbances, limit physical pathomechanisms can occur across different eti-
functioning, lead to psychosocial distress and ologies, has led to a redefinition of classification
thus has a major impact on the quality of life of [6]. Contrary to the original etiology-based clas-
affected patients. Pain management is therefore sification, neuropathic pain should rather be clas-
an important part in the treatment of diabetic neu- sified primarily on the basis of the underlying
ropathy. Several guidelines have been published pathophysiologic mechanisms. The awareness of
for the treatment of neuropathic pain in general these mechanisms that might underlie a patient's
[2, 3] and painful diabetic neuropathy in particu- pain allows the use of drugs that act specifically
lar [4]. However, even with first-line therapeu- on the target structures. This is the concept of an
tics, many affected patients do not receive individualized mechanism-based pain therapy.
sufficient pain relief of their pain, which is Since it is not possible yet to study these
reflected by the high number needed to treat pathomechanisms directly in humans, other
(NNT) ranging from 3.6 for tricyclic antidepres- methods that can provide indirect information
sants to 7.7 for pregabalin [3]. In painful diabetic must be used. The phenotype of a patient, i.e., the
neuropathy, it is estimated that approximately clinical symptoms and signs, are thought to
39% of the patients have never even received a reflect certain underlying mechanisms. Heat
treatment for their pain [5]. In addition, many hyperalgesia, for example, is related to peripheral
clinical trials on new analgesic drugs have failed sensitization [7], whereas pinprick hyperalgesia
in the past. Therefore, an important question that and dynamic mechanical allodynia can be attrib-
has become the focus of pain researchers in uted to central sensitization [8]. Based on this
recent years is: which patients benefit most from idea, efforts have been made to precisely exam-
ine a patient’s phenotype and develop stratifica-
J. Sachau · M. Sendel · R. Baron (*) tion approaches in order to predict treatment
Division of Neurological Pain Research and Therapy, responses.
Department of Neurology, University Hospital
Schleswig-Holstein, Campus Kiel, Kiel, Germany
e-mail: r.baron@neurologie.uni-kiel.de

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 113
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_7
114 J. Sachau et al.

2 The DFNS Protocol ent sensory nerve fiber classes (C-, A-delta,
of Sensory Profiling A-beta-fibers) [9–11].
All assessments are performed in a fixed
2.1 The DFNS Protocol sequence with standardized patient instructions.
of Quantitative Sensory When performed according to protocol, the
Testing DFNS QST protocol shows adequate test–retest
and inter-observer reliability [12] and high homo-
One advance in the effort to adequately define geneity across different centers [13].
somatosensory phenotypes was the standardiza- The DFNS protocol consists of seven tests
tion of Quantitative sensory testing (QST). The including 13 thermal and mechanical parameters
German Research Network on Neuropathic Pain [14, 15]. The protocol should be performed in
(DFNS) developed a standardized test battery two areas for each patient, i.e., in the most
assessing small and large nerve fiber function as affected area and in a normal/unaffected one, in
well as parameters of central processing (Table 1). order to detect deviations from control data and
By using specific techniques to selectively block side-to-side differences [16]. The protocol is dis-
nerve fiber classes, adequate tools have been played in Fig. 1.
identified which asses the entire variety of differ-

Table 1  Quantitative sensory testing (QST) parameters according to the DFNS protocol
Suspected location of
underlying pathophysiological
mechanism QST finding
QST parameter Device Peripheral Central Loss of function Gain of function
CDT Cold detection Thermotest Aδ (cold) Spinothalamic Thermal Thermal
WDT threshold C (warm) hypoesthesia hyperesthesia
Warm detection
threshold
CPT Cold pain threshold Thermotest Aδ, C Spinothalamic Thermal Thermal
HPT Heat pain thresholda hypoalgesia hyperalgesia
PHS Paradoxial heat Thermotest Central Heat sensation
sensation disinhibition to a cold
stimulus
MDT Mechanical Calibrated Aβ Lemniscal Mechanical Mechanical
detection threshold von Frey hypoesthesia hyperesthesia
hairs
VDT Vibration detection Tuning fork Aβ Lemniscal Pallhypoesthesia
threshold
MPT Mechanical pain Pinprick Aδ, C Spinothalamic Pinprick Pinprick
thresholdb stimuli hypoalgesia hyperalgesia
MPS (Suprathreshold) Pinprick Aδ, C Spinothalamic Pinprick Pinprick
mechanical pain stimuli hypoalgesia hyperalgesia
sensitivityb
DMA Dynamic Cotton wisp, Aβ Lemniscal Allodynia
mechanical cotton wool,
allodyniab brush
WUR Wind-up ratio Pinprick Spinothalamic Temporal
stimuli summation
PPT Pressure pain Pressure Aδ, C Spinothalamic Pressure pain Pressure pain
threshold gauge device hypoalgesia hyperalgesia
 Peripheral sensitization
a

 Central sensitization
b
Sensory Profiles and Diabetic Neuropathy 115

a b c
CDT WDT TSL & PHS CPT HPT MDT MPT

intensity steps by a factor of 2


y
ramp for all y y
thermal tests y y y y y y
(1 °C/s) y y
n n
n n n n n n
hot? hot? hot? n n
n
paradoxical
warming heat sensations sensation of touch slightly pricking
cooling during cold? no sensation of touch no pricking

~8 minutes ~4 minutes ~4 minutes

d e f g
MPS ALL WUR VDT PPT
100 100 100
Numerical rating 4
Numerical rating

a b Tuning fork Algometer


10 10 10 3 = b (series) (64 Hz) contact area

Ratio
a (single) 1cm2
1 1 1 2

threshold (kPa)
800 ramp (50kPa/s)

Pressure pain
0.1 0.1 0.1 1
600

Vibratory units (/8)


8
8 16 32 64 128 256 512 CW QT BR trains 1 to 5 6 400
1 2 3 4 5
4 200 1st painful
vibratory pressure
single series of 10 stimuli 2
hyperalgesia allodynia to light touch threshold,
normal sensation to pinprick normal sensation 0 tuning fork
hypoalgesia

~8 minutes ~2 minutes ~2 minutes ~2 minutes

Fig. 1  QST-a battery of sensory tests: figure of methods Stimulus-response functions: mechanical pain sensitivity
(reused from Rolke et  al. [15]). The standardized QST (MPS) for pinprick stimuli, and dynamic mechanical allo-
protocol assesses 13 parameters in seven test procedures dynia (ALL) assess A-delta mediated sensitivity to sharp
(a–g). All procedures are presented including a time frame stimuli (pinprick), and also A-beta fiber mediated pain
for testing over one area. (a) Thermal testing comprises sensitivity to stroking light touch (CW cotton wisp, QT
detection and pain thresholds for cold, warm, or hot stim- cotton wool tip, BR brush). (e) Wind-up ratio (WUR)
uli (C- and A-delta fiber mediated): cold detection thresh- compares the numerical ratings within five trains of a
old (CDT); warm detection threshold (WDT); number of single pinprick stimulus (a) with a series (b) of 10 repeti-
paradoxical heat sensations (PHS) during the thermal sen- tive pinprick stimuli to calculate WUR as the ratio: b/a. (f)
sory limen procedure (TSL) for alternating warm and cold Vibration detection threshold (VDT) tests for A-beta fiber
stimuli; cold pain threshold (CPT); heat pain threshold function using a Rydel-Seiffer 64  Hz tuning fork. (g)
(HPT). (b) Mechanical detection threshold (MDT) tests Pressure pain threshold (PPT) is the only test for deep
for A-beta fiber function using von Frey-filaments. (c) pain sensitivity, most probably mediated by muscle C-
Mechanical pain threshold (MPT) tests for A-delta fiber and A-delta fibers. For details regarding the testing proce-
mediated hyper- or hypoalgesia to pinprick stimuli. (d) dures also see Sect. 2

2.1.1 Small Fiber Function s­ensations (PHS) is captured. Thermal thresh-


(C-/A-­delta): Thermal Thresholds olds allow assessing small fiber function or their
Thermal thresholds are assessed using a ther- corresponding central pathways. PHS, however,
mode of a defined size with a baseline tempera- can also occur in case of disturbed central
ture of 32 °C.  Temperature ramps are used to processing.
determine thermal detection (cold detection Heat hyperalgesia is predominantly caused by
threshold, CDT; warm detection threshold, peripheral sensitization [7]. The heat sensitive
WDT) as well as pain thresholds (cold pain ion channel TRPV1 can be sensitized through
threshold, CPT; heat pain threshold, HPT) different mechanisms, e.g., inflammatory media-
using the method of levels. Furthermore, the tors, which causes the heat pain threshold to drop
ability to discriminate temperatures (thermal [17]. The same is true for TRPA1, whose sensiti-
sensory limen, TSL) including paradoxical heat zation causes cold hyperalgesia [18].
116 J. Sachau et al.

2.1.2 A-Beta-Fiber-Function: able pain). In total, 35 pinprick stimuli are applied


Mechanical Detection to each testing site.
Thresholds Pinprick stimuli are alternated with normally
The mechanical detection threshold (MDT) for innocuous tactile stimuli (cotton wisp, Q-tip, soft
touch is determined using modified von Frey brush) to test for dynamic mechanic allodynia
hairs (0.25 to 512 mN) [19]. The patient’s detec- (DMA).
tion threshold is identified using the method of Additionally, subjects are asked to rate a sin-
limits, applying the stimuli in ascending and gle pinprick stimulus (256 mN or 128 mN when
descending order. testing in the face) as well as ten consecutive
The mechanical detection threshold for vibra- stimuli applied as one per second in a 1 cm2 area.
tion (vibration detection threshold, VDT) is a dis- To determine the WUR the mean rating of five
appearance threshold. A standard Rydel-Seiffer series of stimuli is divided by the mean rating of
tuning fork (64 Hz, 8/8 scale) is administered at a five single stimuli. The WUR correlates to an
bone protrusion three times. increase in excitability of spinal neurons, and is
therefore an indication of the status of spinal pro-
2.1.3 A-Delta Fiber Function cessing [20].
and Central Sensitization: For determining the pressure pain threshold
Pinpricks and Pressure Pain (PPT), a pressure algometer is pressed to a mus-
Threshold, Dynamic Mechanic cle with a force up to 20 kg/cm2 and a ramp of 0.5
Allodynia kg/cm2 per second. PPT is the only QST param-
Weighted pinpricks (8–512 mN) are used to eter used to assess deep somatic sensitivity.
determine the mechanical pain threshold (MPT), Abnormal MPT, WUR, MPS, and PPT can
mechanical pain sensitivity (MPS), and the wind- indicate A-delta fiber damage (or their corre-
­up ratio (WUR). sponding central pathways). In addition, mechan-
Pinprick hyperalgesia is evaluated by both the ical hyperalgesia and mechanical allodynia are
MPT and MPS. The method of limits is used for surrogates of central sensitization [17].
the assessment of MPT.  The patient is asked to
rate each pinprick as “dull” or “sharp.” Pinpricks 2.1.4 Z-transformation
are administered in ascending and descending and Reference Values
order of sharpness. A single patient’s QST values can be compared
MPS is based on a stimulus-response func- with reference values regardless of the different
tion. Subjects have to rate the pain intensity of kinds of measurement. This is possible due to
each stimulus on a numeric rating scale (NRS, 0 Z-transformation of the values (except for PHS
meaning no pain, 100 meaning the worst imagin- and DMA, see below) [21]:

Z − score = ( value of the single patient − mean of controls ) / standard deviation of controls.

Therefore, all QST parameters can be depicted above zero indicate a gain of function, whereas
as standard normal distributions with a mean of Z-scores below zero indicate a loss. For this
zero. PHS and DMA, that are absent under nor- ­purpose, the algebraic sign is changed accord-
mal conditions, are given with original values. ingly in most of the parameters so all values indi-
For the majority parameters, which are not cating loss of function show a Z-score below zero
normally distributed (CDT, WDT, TSL, MDT, and vice versa [15]. This makes it possible to eas-
MPT, MPS, DMA, WUR, PPT), a logarithmic ily grasp loss and gain of sensory function at a
transformation is performed beforehand. Z-scores glance.
Sensory Profiles and Diabetic Neuropathy 117

As QST values differ significantly across body Despite these limitations, QST has some
regions, between men and women and between important advantages. QST enables to determine
younger and older people, each body area a comprehensive somatosensory profile in a sin-
requires different gender and age specific refer- gle test session. This includes the function of
ence values [14, 22]. Therefore, reference values both large and small fibers, which cannot be
were established for different age groups, sepa- assessed using conventional methods of electro-
rately for men and women as well as for specific physiology. Unlike other methods used for the
body areas including the dorsum of the foot and assessment of small fiber function, QST cannot
hand [14], the face and the trunk [23]. Reference only detect loss of function, but is also highly
data are given as means and 95% confidence sensitive to positive signs, i.e. gain of function
intervals (mean ± 1.96 SD), thus values above [15]. Thus, QST can be used as a potential bio-
+1.96 indicate an abnormal gain and values marker for the diagnosis of (small fiber) neuropa-
below –1.96 an abnormal loss of function. thies, disease progression and prediction of
treatment response [25].
2.1.5 Advantages and Disadvantages
of QST
There are several disadvantages that should be 2.2 Sensory Profiling According
kept in mind when performing and evaluating to the DFNS QST Protocol
QST according to the DFNS protocol. First of
all, and perhaps most importantly, QST is a psy- 2.2.1 The DFNS QST Protocol
chophysical measure and therefore requires Theoretically, the detection of 26 different abnor-
patient compliance. Any aspect that hinders malities is possible considering the entirety of 13
patients’ understanding of the instructions like parameters of the DFNS QST protocol. As DMA
cognitive deficits or a language barrier therefore and PHS are absent in healthy subjects, an abnor-
must be excluded prior to testing. In addition, it mal loss of function is not possible. Furthermore,
is important to perform QST according to the it has to be noted that the upper confidence limits
protocol using the pre-defined instructions to of the thermal pain thresholds (CPT, HPT) as
guarantee reliable results. As mentioned above, well as the lower confidence thresholds of MDT
reference values are required for each area tested and VDT are close to the limits of possible attain-
to classify results as normal or abnormal. When able values. This limits the detection of thermal
there are no available reference data, comparison hypoalgesia and mechanical hyperesthesia. As
to the contralateral site should be used to judge QST values significantly correlate across body
sensory function of the tested area [14]. All data sites in healthy individuals, comparing absolute
in the DFNS data base have been obtained from QST values to the contralateral site may provide
Caucasian subjects. Thus a higher variability of insight into abnormalities that otherwise could be
values can be expected in a cohort of healthy missed [14].
subjects of different ethnicity [24]. To avoid tis- All things considered, 20 different phenomena
sue damage, the range of available values is lim- of hypo- or hyperesthesia can be detected.
ited. For some of the parameters the range of The simplest approach of profiling patients is
normal values overlaps with these cut-off values the description of existence or absence of one
so that the definition of abnormal values is not distinct symptom, e.g., allodynia. Considering all
possible. This, to some extent, limits the assess- parameters of the DFNS QST protocol, there are
ment of sensory loss, especially in older patients many different profiles that can theoretically
[15]. In addition, QST cannot determine the occur, although it is not always appropriate or
exact location of the lesion or disease, thus a dif- necessary to consider all of them [26].
ferentiation between a damage of peripheral The two most commonly described sensory
nerve fibers or their correlating central pathways profiles are the so-called irritable nociceptor (IN)
is not possible. phenotype, characterized by hypersensitivity, and
118 J. Sachau et al.

the “deafferentation“ or “non-irritable nocicep- tion sensation was diminished regardless of pain.
tor” (NIN) phenotype characterized by loss of Although children with type 1 diabetes did show
function. Those phenotypes will be described a higher MPS, this parameter generally seems to
further below. play a smaller role in diabetic neuropathy [22].
Only a small percentage of patients present a loss
of function. This, again, is more pronounced in
2.3 Sensory Profiling in Diabetic patients with neuropathic pain [30, 31]. DMA is
Neuropathy rare in patients with diabetic neuropathy, it was
only found in a small percentage of patients with
Single parameters of QST, especially thermal neuropathic pain (17% of patients with mild neu-
parameters and vibration, have been used in stud- ropathic pain, 14% of patients with moderate to
ies regarding diabetic neuropathy for quite some severe neuropathic pain [30]; 7.6% of all patients
time. with neuropathic pain [31]).
The DFNS protocol, however, provides an Accordingly, patients with diabetic neuropa-
opportunity to compile a complete sensory pro- thy seem to be more likely to fit the deafferenta-
file in a structured way. tion profile (e.g., 53.8% of patients with
In 2012 Blankenburg et al. used QST to screen neuropathic pain [31]). The severity of loss cor-
for diabetic neuropathy in children with type 1 relates with the presence and severity of neuro-
diabetes and found subclinical large and small pathic pain, neuropathy in general and disability
fiber neuropathies in almost 50% of them [27]. [31]. When differentiating between polyneu-
Many studies using QST to stratify patients ropathy patients with and without pain, those
especially put an emphasis on finding differences with pain tend to show more significant loss of
between patients with painful and painless dia- function, while the significance correlates with
betic neuropathy [28]. In two studies patients the severity of neuropathy in general [31]. Gain
were divided into three groups: patients without of function seems to be much rarer. Only a
neuropathic pain, with mild neuropathic pain, small percentage (4.9–7.1% of patients with
and those with moderate to severe neuropathic mild neuropathic pain, 7.1–7.5% of patients
pain. with moderate to severe neuropathic pain [30];
Abnormal QST-values, most commonly rep- 14.6% in total [31]) of patients display the
resenting loss of function, can be found in most characteristics of the irritable nociceptor (IN)
patients with diabetic neuropathy, although they profile. Patients with the IN profile show lower
are more common when patients report pain [29, neuropathy disability scores and a shorter dura-
30]. Both large and small fiber functions are tion of pain.
affected [27, 30]. DMA and PHS as symptoms of aberrant cen-
When looking at thermal thresholds, generally tral processing seem to occur more often in
lower Z-values can be found in patients with dia- patients with more severe pain [31].
betic neuropathy than in healthy controls [30],
regardless of whether patients suffer from pain or
not [29]. 3 Patient Stratification
Significant gain was generally rare. An excep- for Mechanism-Based
tion was heat hyperalgesia in the study assessing Treatment
children with type 1 diabetes [27]. PHS is found
in all groups of patients with diabetic neuropathy. 3.1 Clustering Approach
The lowest Z-scores are present in patients with
moderate to severe pain [30, 31]. Regarding In 2016, the DFNS has developed a clustering
MDT there is also a high frequency of loss, which approach to stratify neuropathic pain patients
is again most pronounced in patients with painful into groups of similar clinical signs based on
neuropathy. In all studies considered VDT, vibra- their QST sensory profile [32]. In total, 902
Sensory Profiles and Diabetic Neuropathy 119

patients with peripheral neuropathic pain of dif- Cluster 1 was characterized by a loss of small
ferent etiology were included in this study, more and large nerve fiber function, i.e., hypoesthesia
than half of them (57%) suffered from painful to thermal and mechanical stimuli, as well as par-
polyneuropathy. Three clusters, each of them adoxical heat sensation. At the mechanism level,
characterized by similar QST profiles, i.e. pre- this cluster resembles the deafferentation caused
sumably similar pathophysiological mechanisms, by a compression nerve block. Cluster 2 showed
were identified: cluster 1 (sensory loss), cluster 2 intact nerve fiber function combined with thermal
(thermal hyperalgesia), and cluster 3 (mechanical hyperalgesia to heat and cold. This cluster was
hyperalgesia) (Fig.  2, Table  2). Interestingly, found to reflect the human surrogate model of
these three clusters could also be identified in peripheral sensitization induced by enhanced acti-
human surrogate models of neuropathic pain, vation of TRPV1 receptors. Finally, cluster 3 was
supporting the mechanistical relevance of these characterized by a loss of small nerve fiber func-
sensory phenotypes [33]. tion in combination with hyperalgesia to blunt

Sensory loss
Thermal hyperalgesia 3
3 10
Mechanical hyperalgesia
n=902
gain of function

Pain rating (NRS)

PHS reports
2 2

1 1
.3
Z-score

0 0 0
DMA PHS

–1
loss of function

–2

–3

CDT WDT TSL CPT HPT PPT MPT MPS WUR MDT VDT

QST parameter

Fig. 2  QST profiles of the 3-cluster solution (adapted dynamic mechanical allodynia (DMA) on a logarithmic
from Baron et al. [32]). Sensory profiles of the three clus- scale (0–100) and frequency of paradoxical heat sensation
ters presented as mean Z-scores ± 95% confidence inter- (PHS) (0–3). Blue symbols: cluster 1 “sensory loss”
val (n = 902). Note that Z-transformation eliminates (42%). Red symbols: cluster 2 “thermal hyperalgesia”
differences due to test site, sex, and age. Positive Z-scores (33%). Yellow symbols: cluster 3 “mechanical hyperalge-
indicate positive sensory signs (hyperalgesia), whereas sia” (24%). CDT cold detection threshold, CPT cold pain
negative Z-values indicate negative sensory signs (hypo- threshold, HPT heat pain threshold, MDT mechanical
esthesia and hypoalgesia). Dashed lines: 95% confidence detection threshold, MPS mechanical pain sensitivity,
interval for healthy subjects (−1.96 < z < +1.96). Note that MPT mechanical pain threshold, NRS numerical rating
if the mean of a cluster is within the shaded area, this does scale, PPT pressure pain threshold, QST quantitative sen-
not imply that it does not differ from a healthy cohort. sory testing, TSL thermal sensory limen, VDT vibration
Values are significantly different from those of healthy detection threshold, WDT warm detection threshold,
subjects, if their 95% confidence interval does not cross WUR wind-up ratio
the zero line. Insets show numeric pain ratings for
120 J. Sachau et al.

Table 2  Characteristics of the three QST clusters


Postulated
Cluster QST findings pathomechanism Therapeutical implications
Sensory loss Touch, vibration, Loss of small and ++ Antidepressants, opioids
temperature, pain large fiber function
Paradoxical heat + Gabapentinoids, sodium channel
sensation blocker
Thermal None Peripheral ++ Sodium channel blocker
hyperalgesia sensitization
Heat, cold hyperalgesia + Antidepressants, gabapentinoids,
capsaicin patches, opioids, botulinum
toxin (NSAID)
Mechanical In particular warm and Loss of small fiber ++ Gabapentinoids, sodium channel
hyperalgesia cold hypoesthesia function blocker
Pressure, pinprick Central sensitization + Antidepressants, opioids, NMDA
hyperalgesia dynamic antagonists
mechanical allodynia
loss of function, gain of function, NSAID non-steroidal anti-inflammatory drug, NMDA N-methyl-D-aspartate

pressure and pinprick as well as dynamic mechan- with the required number of patients for each
ical allodynia, patterns that were also found in condition can be found in the paper. For example,
surrogate models of central sensitization. in order to detect an adequate subgroup of
These three clusters were detected in each of patients allocated to the sensory loss cluster in a
the included disease etiologies, i.e., polyneuropa- cross-over study on painful diabetic neuropathy
thy, radiculopathy, peripheral nerve injury, and with an estimated effect size of 0.5, about 41
postherpetic neuralgia, though with different fre- patients need to be screened. In contrast, the
quencies. In a second step, an algorithm with detection of a mechanical hyperalgesia subgroup
both a deterministic and a probabilistic version requires more than twice as many patients (n =
was developed to allocate individual patients to 103) in the same setting.
the three pre-defined clusters [34]. Using the The identification of specific patient clusters
deterministic approach, each patient is assigned is a first step towards a mechanism-based ther-
to exactly one cluster, whereas the probabilistic apy. The Committee for Medicinal Products for
approach allows the allocation of one patient to Human Use (CHMP) of the European Medicines
more than one cluster. By doing so, patients with Agency (EMA) has therefore acknowledged the
painful diabetic polyneuropathy were mostly QST-based sensory subgrouping an adequate
assigned to the sensory loss cluster (64% for the stratification tool for identification of specific
deterministic approach; 82% for the probabilistic sensory phenotypes of patients in clinical trials
approach), followed by the mechanical hyperal- on neuropathic pain. To overcome the well-­
gesia cluster (19%; 75%) and least frequently the known limitations of QST, i.e., the time-­
thermal hyperalgesia cluster (13%; 33%). This consuming protocol and expensive equipment,
observation is in agreement with the results of several research groups have concentrated on the
Themistocleous et al. [30]. Finally, Vollert et al. development of easy-to-use bedside testing pro-
proposed recommendations on the sample size, cedure that can be easily applied in both large
i.e., the number of patients wo need to be screened clinical trials and clinical practice [35, 36].
to find a subgroup that is large enough to conduct Recently, it was shown that a combination of
a clinical trial (cross-over or parallel design) with only five simple bedside parameters is necessary
a power of 80%, an alpha-level of 0.05 and an for the correct allocation to the three pre-defined
effect size of 0.3, 0.5, or 0.7 [34]. A detailed table QST clusters [37].
Sensory Profiles and Diabetic Neuropathy 121

3.2 Sensory Testing as Predictor another study, this proportion was even higher
of Treatment Response suggesting that about 50% of all patients with
painful diabetic neuropathy are characterized by
In the last years, several studies have been pub- intact sensory function and would therefore ben-
lished that support the phenotype-based stratifi- efit from a therapy with oxcarbazepine [44].
cation as a possible approach to identify However, other results imply a much smaller
responders in clinical trials on neuropathic pain proportion of this subgroup [30], underscoring
in general, and diabetic neuropathy in particular. that more stratification studies are needed to
Demant et al. performed a placebo-controlled, define the exact phenotype distribution in
cross-over trial to investigate the efficacy of patients with diabetic neuropathy. Nevertheless,
oxcarbazepine, an unselective sodium channel this study nicely shows that the idea of a QST-
blocker, in patients with peripheral neuropathic based subgrouping can improve the treatment of
pain of different etiology [38]. As one of the first peripheral neuropathic pain in general. But there
studies with an a priori stratification, patients are also promising studies for diabetic neuropa-
were prospectively subgrouped according to their thy supporting the phenotype-based stratifica-
QST profile into a NIN (n = 52) and a IN type (n tion as predictive biomarker for treatment
= 31) based on a previously published LoGA response.
classification [16]. Even though this classifica- TRPA1 has been shown to induce cold and
tion does not correspond completely to the three-­ also mechanical hyperalgesia in animal models
cluster sorting algorithm of Baron et  al. [32], of (diabetic) neuropathic pain [45]. In a double-
both approaches share some similarities. The IN blind, placebo-controlled Phase 2a study on pain-
type is characterized by an intact sensory func- ful diabetic neuropathy, the selective TRPA1
tion for cold and warm combined with thermal antagonist GRC 17536 was shown to be effective
and/or mechanical hyperalgesia and hence corre- in a subgroup of patients with moderate to severe
sponds to the thermal hyperalgesia cluster 2, pain and preserved sensory function (similar to
while the NIN is characterized by profound the irritable nociceptor type) as detected via ret-
­sensory loss, although the area can be painful rospective QST-based stratification [44].
nevertheless [39]. Another option to stratify patients based on
The upregulation or increased expression and their phenotype is the assessment of conditioned
activation of TRPV1 [40], voltage-gated sodium pain modulation (CPM). This “pain inhibits pain
channels [41, 42] and voltage-gated calcium phenomenon” describes the reduction in pain
channels [43] are proposed mechanisms respon- intensity of a first painful stimulus (test stimulus)
sible for irritable nociceptors [41, 42]. by a second painful stimulus (conditioning stim-
Consequently, drugs like oxcarbazepine acting as ulus). Based on the hypothesis that certain drugs
sodium channel blockers ought to be more effec- that enhance descending inhibitory pathways
tive in patients with an increased expression and would be more effective in patients with
activation of sodium channels. insufficient CPM effect, Yarnitsky et  al. con-
­
This theoretical assumption was confirmed in ducted an exploratory study in patients with pain-
the study by Demant et al. [38]. While the NNT ful diabetic neuropathy [46]. In total, 30 patients
for a 50% pain reduction was as high as 13 in the received placebo followed by duloxetine, a com-
NIN type, it decreased to 3.9 in the IN subgroup. bined serotonin/norepinephrine reuptake inhibi-
Therefore, oxcarbazepine may be considered as tor. Interestingly, a less efficient CPM effect
therapeutic option in a certain subgroup of before the treatment period was associated with
patients. With respect to pain in diabetic neurop- higher drug efficacy. Therefore, the authors con-
athy, this seems to be the case in about one third cluded that phenotype-based stratification using a
of all patients when applying the probabilistic simple CPM protocol may be a predictor for
approach of the DFNS sorting algorithm. In treatment response to duloxetine.
122 J. Sachau et al.

3.3 Sensory Profiling Using with diabetic neuropathy exhibited the IN profile
Patient Reported Outcome in this study. The profile indicating deafferenta-
Measures tion pain was only second most common. There
are several possible explanations for this result: It
As described above, QST does have its limits is possible that questionnaires are less suited to
[47]. While its results depict evoked pain very assess negative symptoms like numbness, mak-
well, spontaneous pain cannot be assessed. Apart ing positive symptoms overrepresented.
from that, QST results do not necessarily repre- Likewise, evoked pain seems to be reported by
sent the symptoms patients actually feel. more patients in comparison to the number of
Several studies have used questionnaires, patients showing DMA in QST [30, 31]. In addi-
originally designed to screen for neuropathic tion, the patients included in this study overall
pain components [48], to stratify patients consid- were in an earlier stage of disease and therefore
ering the pattern of sensory abnormalities displayed the IR profile more often, similar to
reported by themselves. The painDETECT ques- what is the case in QST profiles in animal models
tionnaire as well as the Neuropathic Pain [55]. While symptoms and signs of gain seem to
Symptom Inventory (NPSI) have been able to be more common in early disease stages and in
successfully identify different subgroups [49]. case of small fiber damage, a sensory profile of
The Standardized Evaluation of Pain (StEP), a loss rather occurs with longer disease duration
combination of interview questions and physical [55, 56]. In animal models, however, there are
examination, was used to identify eight different other causes as well that may explain the lack of
patient clusters. One cluster characterized by agreement in the QST profile between animal
symptoms of loss was specific to diabetic neu- and patient. Streptozotocin-induced diabetes,
ropathy [50]. which is the most commonly used model, does
In a large study using the painDETECT ques- not match the characteristics of diabetes mellitus
tionnaire, patients with diabetic neuropathy most in humans exactly [57]. Furthermore, it can be
commonly exhibited a sensory profile character- argued that the assessment of sensory loss in ani-
ized by prominent numbness. A considerable mals is not as practicable as it is in humans,
percentage of patients also described suffering meaning that symptoms of loss may be underrep-
from burning pain and paresthesia. Diabetic neu- resented in animal studies [58].
ropathy patients less commonly suffered from Recently, a questionnaire-based stratification
pain attacks and burning pain in combination approach has been used by Bouhassira and col-
with paresthesia and allodynia. Pressure hyper- leagues who used the NPSI to identify clusters in
algesia in combination with allodynia was least a large cohort of 628 patients with probable or
common [51]. definite neuropathic pain [59]. First, they identi-
A study using the NPSI [52] found quite simi- fied three subgroups that were characterized by
lar results as paresthesia and burning were the pain different symptom patterns: the deep pain cluster
descriptors used most often by patients with dia- (higher scores for squeezing and pressure pain),
betic neuropathy. Paroxysmal pain, evoked pain, the evoked pain cluster (higher evoked pain
and deep pain were reported less commonly [53]. scores and low deep pain scores), and the pin-
The pain PREDICT questionnaire [54] was pointed pain cluster (higher scores for tingling
especially developed to predict treatment and pins and needles plus moderate burning and
response as well as assess different neuropathic paroxysmal scores). Afterwards they developed
and nociceptive symptoms and categorize an algorithm to assign patients to one of these
patients into different subgroups. The commonly clusters. Finally, they used this approach to per-
used IN and NIN profiles as well as a third one form a post-hoc analysis of two placebo-­
“pain attacks with nociceptive component” were controlled studies in order to predict treatment
used within this approach as well. Contrary to the response to subcutaneous botulinum toxin A
other above-­ mentioned results, most patients injections. Interestingly, the evoked pain cluster
Sensory Profiles and Diabetic Neuropathy 123

as well as the deep pain cluster showed a signifi- 6. von Hehn CA, Baron R, Woolf CJ.  Deconstructing
cant treatment response, whereas the pinpointed the neuropathic pain phenotype to reveal neural
mechanisms. Neuron. 2012;73(4):638–52. https://doi.
pain cluster failed to show a significant effect. org/10.1016/j.neuron.2012.02.008.
While patients with diabetic neuropathy were a 7. LaMotte RH, Thalhammer JG, Torebjörk HE,
minority in this study, a similar algorithm could Robinson CJ. Peripheral neural mechanisms of cuta-
be applied in different cohorts in the future. neous hyperalgesia following mild injury by heat. J
Neurosci. 1982;2(6):765–81.
Overall, phenotypic-based stratification to 8. LaMotte RH, Shain CN, Simone DA, Tsai
predict treatment response has become more and EF.  Neurogenic hyperalgesia: psychophysical stud-
more popular in recent years. The results of first ies of underlying mechanisms. J Neurophysiol.
clinical trials in which a prospective subgrouping 1991;66(1):190–211. https://doi.org/10.1152/
jn.1991.66.1.190.
was performed have shown promising results, 9. Fruhstorfer H.  Thermal sensibility changes dur-
indicating that this is an appropriate approach to ing ischemic nerve block. Pain. 1984;20(4):355–61.
realize the concept of an individualized https://doi.org/10.1016/0304-­3959(84)90112-­X.
mechanism-­based therapy. In future, the use of 10. Yarnitsky D, Ochoa JL.  Differential effect of
compression-­ ischaemia block on warm sensation
simple bedside tools combined with validated and heat-induced pain. Brain. 1991;114(2):907–13.
patient reported outcome measures may help to https://doi.org/10.1093/brain/114.2.907.
apply a priori sensory stratification also in large 11. Ziegler EA, Magerl W, Meyer RA, Treede
clinical trials. This will hopefully result in RD.  Secondary hyperalgesia to punctate mechani-
cal stimuli. Central sensitization to A-fibre nocicep-
improved treatment strategies for neuropathic tor input. Brain. 1999;122(12):2245–57. https://doi.
pain in general and painful diabetic neuropathy in org/10.1093/brain/122.12.2245.
particular. 12. Geber C, et  al. Test-retest and interobserver reli-
ability of quantitative sensory testing according to
the protocol of the German Research Network on
Acknowledgement We would like to thank Dilara Neuropathic Pain (DFNS): a multi-centre study.
Kersebaum for her excellent help in language editing. Pain. 2011;152(3):548–56. https://doi.org/10.1016/j.
pain.2010.11.013.
13. Vollert J, et  al. Quantitative sensory testing using
DFNS protocol in Europe: an evaluation of het-
References erogeneity across multiple centers in patients with
peripheral neuropathic pain and healthy subjects.
1. Sloan G, et  al. A new look at painful diabetic neu- Pain. 2016;157(3):750–8. https://doi.org/10.1097/j.
ropathy. Diabetes Res Clin Pract. 2018;144:177–91. pain.0000000000000433.
https://doi.org/10.1016/j.diabres.2018.08.020. 14. Rolke R, et  al. Quantitative sensory testing in the
2. Attal N, et  al. EFNS guidelines on the pharmaco- German Research Network on Neuropathic Pain
logical treatment of neuropathic pain: 2010 revi- (DFNS): standardized protocol and reference values.
sion. Eur J Neurol. 2010;17(9):1113. https://doi. Pain. 2006;123(3):231–43. https://doi.org/10.1016/j.
org/10.1111/j.1468-­1331.2010.02999.x. pain.2006.01.041.
3. Finnerup NB, et al. Pharmacotherapy for neuropathic 15. Rolke R, et  al. Quantitative sensory testing: a
pain in adults: a systematic review and meta-­analysis. comprehensive protocol for clinical trials. Eur J
Lancet Neurol. 2015;14(2):162–73. https://doi. Pain. 2006;10(1):77–88. https://doi.org/10.1016/j.
org/10.1016/S1474-­4422(14)70251-­0. ejpain.2005.02.003.
4. Bril V, et al. Evidence-based guideline: treatment of 16. Maier C, et  al. Quantitative sensory testing in the
painful diabetic neuropathy–report of the American German Research Network on Neuropathic Pain
Association of Neuromuscular and Electrodiagnostic (DFNS): somatosensory abnormalities in 1236
Medicine, the American Academy of Neurology, patients with different neuropathic pain syndromes.
and the American Academy of Physical Medicine Pain. 2010;150(3):439–50. https://doi.org/10.1016/j.
& Rehabilitation. Muscle Nerve. 2011;43(6):910–7. pain.2010.05.002.
https://doi.org/10.1002/mus.22092. 17. Treede R-D. Chapter 1 pain and hyperalgesia: defini-
5. Daousi C, MacFarlane IA, Woodward A, Nurmikko tions and theories. Handb Clin Neurol. 2006;81:3–10.
TJ, Bundred PE, Benbow SJ.  Chronic painful https://doi.org/10.1016/S0072-­9752(06)80005-­9.
peripheral neuropathy in an urban community: a 18. Obata K, et  al. TRPA1 induced in sensory neurons
controlled comparison of people with and without contributes to cold hyperalgesia after inflammation
diabetes. Diabet Med. 2004;21(9):976–82. https://doi. and nerve injury. J Clin Invest. 2005;115(9):2393–
org/10.1111/j.1464-­5491.2004.01271.x. 401. https://doi.org/10.1172/JCI25437.
124 J. Sachau et al.

19. Fruhstorfer H, Gross W, Selbmann O. von Frey sensory profiles. Pain. 2017;158(2):261–72. https://
hairs: new materials for a new design. Eur J doi.org/10.1097/j.pain.0000000000000753.
Pain. 2001;5(3):341–2. https://doi.org/10.1053/ 33. Vollert J, et  al. Pathophysiological mechanisms of
eujp.2001.0250. neuropathic pain: comparison of sensory phenotypes
20. Herrero JF, Laird JM, López-García JA. Wind-up of in patients and human surrogate pain models. Pain.
spinal cord neurones and pain sensation: much ado 2018;159(6):1090–102. https://doi.org/10.1097/j.
about something? Prog Neurobiol. 2000;61(2):169– pain.0000000000001190.
203. https://doi.org/10.1016/s0301-­0082(99)00051-­9. 34. Vollert J, et  al. Stratifying patients with peripheral
21. Glass GV, Stanley JX. Statistical methods in education neuropathic pain based on sensory profiles: algo-
and psychology. Englewood Cliffs: Prentice-­Hall; 1970. rithm and sample size recommendations. Pain.
22. Blankenburg M, et  al. Reference values for quanti- 2017;158(8):1446–55. https://doi.org/10.1097/j.
tative sensory testing in children and adolescents: pain.0000000000000935.
developmental and gender differences of somatosen- 35. Buliteanu A, et  al. Validation of a bedside quantita-
sory perception. Pain. 2010;149(1):76–88. https://doi. tive sensory testing (QST) protocol in chronic neu-
org/10.1016/j.pain.2010.01.011. ropathic pain. J Pain. 2018;19(3):S52. https://doi.
23. Pfau DB, et  al. Quantitative sensory testing in the org/10.1016/j.jpain.2017.12.123.
German Research Network on Neuropathic Pain 36. Zhu GC, et al. Concurrent validity of a low-cost and time-
(DFNS): reference data for the trunk and applica- efficient clinical sensory test battery to evaluate somato-
tion in patients with chronic postherpetic neuralgia. sensory dysfunction. Eur J Pain. 2019;23(10):1826–38.
Pain. 2014;155(5):1002–15. https://doi.org/10.1016/j. https://doi.org/10.1002/ejp.1456.
pain.2014.02.004. 37. Reimer M, et  al. Sensory bedside testing: a simple
24. Gierthmühlen J, et  al. Who is healthy? Aspects stratification approach for sensory phenotyping.
to consider when including healthy volunteers in Pain Rep. 2020;5(3):e820. https://doi.org/10.1097/
QST--based studies-a consensus statement by the PR9.0000000000000820.
EUROPAIN and NEUROPAIN consortia. Pain. 38. Demant DT, et  al. The effect of oxcarbazepine in
2015;156(11):2203–11. https://doi.org/10.1097/j. peripheral neuropathic pain depends on pain pheno-
pain.0000000000000227. type: a randomised, double-blind, placebo-controlled
25. Smith SM, et al. The potential role of sensory testing, phenotype-stratified study. Pain. 2014;155(11):2263–
skin biopsy, and functional brain imaging as biomark- 73. https://doi.org/10.1016/j.pain.2014.08.014.
ers in chronic pain clinical trials: IMMPACT con- 39. Fields HL, Rowbotham M, Baron R.  Postherpetic
siderations. J Pain. 2017;18(7):757–77. https://doi. neuralgia: irritable nociceptors and deafferenta-
org/10.1016/j.jpain.2017.02.429. tion. Neurobiol Dis. 1998;5(4):209–27. https://doi.
26. Baron R, Förster M, Binder A. Subgrouping of patients org/10.1006/nbdi.1998.0204.
with neuropathic pain according to pain-related sen- 40. Suh Y-G, Oh U.  Activation and activators of
sory abnormalities: a first step to a stratified treatment TRPV1 and their pharmaceutical implication. Curr
approach. Lancet Neurol. 2012;11(11):999–1005. Pharm Des. 2005;11(21):2687–98. https://doi.
https://doi.org/10.1016/S1474-­4422(12)70189-­8. org/10.2174/1381612054546789.
27. Blankenburg M, et  al. Childhood diabetic neuropa- 41. Devor M. Sodium channels and mechanisms of neu-
thy: functional impairment and non-invasive screen- ropathic pain. J Pain. 2006;7(1):3–12. https://doi.
ing assessment. Diabet Med. 2012;29(11):1425–32. org/10.1016/j.jpain.2005.09.006.
https://doi.org/10.1111/j.1464-­5491.2012.03685.x. 42. Dib-Hajj SD, Cummins TR, Black JA, Waxman
28. Shillo P, et  al. Painful and painless diabetic neu- SG.  Sodium channels in normal and pathological
ropathies: what is the difference? Curr Diab Rep. pain. Annu Rev Neurosci. 2010;33:325–47. https://
2019;19(6):32. https://doi.org/10.1007/s11892-­019-­ doi.org/10.1146/annurev-­neuro-­060909-­153234.
1150-­5. 43. Bourinet E, Altier C, Hildebrand ME, Trang T, Salter
29. Krämer HH, Rolke R, Bickel A, Birklein F. Thermal MW, Zamponi GW. Calcium-permeable ion channels
thresholds predict painfulness of diabetic neuropa- in pain signaling. Physiol Rev. 2014;94(1):81–140.
thies. Diabetes Care. 2004;27(10):2386–91. https:// https://doi.org/10.1152/physrev.00023.2013.
doi.org/10.2337/diacare.27.10.2386. 44. Tandon M et  al. A simple algorithm to identify
30. Themistocleous AC, et  al. The pain in neuropa- likely responders to GRC 17356 in patients of pain-
thy study (PiNS): a cross-sectional observational ful diabetic peripheral neuropathy using sensory
study determining the somatosensory phenotype mapping
of painful and painless diabetic neuropathy. Pain. 45. de Araujo DSM, Nassini R, Geppetti P, De Logu
2016;157(5):1132–45. https://doi.org/10.1097/j. F. TRPA1 as a therapeutic target for nociceptive pain.
pain.0000000000000491. Expert Opin Ther Targets. 2020;24(10):997–1008.
31. Raputova J, et  al. Sensory phenotype and risk fac- https://doi.org/10.1080/14728222.2020.1815191.
tors for painful diabetic neuropathy: a cross-sectional 46. Yarnitsky D, Granot M, Nahman-Averbuch H,
observational study. Pain. 2017;158(12):2340–53. Khamaisi M, Granovsky Y. Conditioned pain modu-
https://doi.org/10.1097/j.pain.0000000000001034. lation predicts duloxetine efficacy in painful diabetic
32. Baron R, et  al. Peripheral neuropathic pain: a neuropathy. Pain. 2012;153(6):1193–8. https://doi.
mechanism-­ related organizing principle based on org/10.1016/j.pain.2012.02.021.
Sensory Profiles and Diabetic Neuropathy 125

47. Sachau J, Baron R. Neuropathic pain therapy: a puzzle 54. Tölle TR, et  al. Pain predict: first interim data from
of different approaches to stratify patients. Pain. 2020. the development of a new patient-reported pain
https://doi.org/10.1097/j.pain.0000000000002120 questionnaire to predict treatment response using
48. Freynhagen R, Baron R, Gockel U, Tölle TR.  Pain sensory symptom profiles. Curr Med Res Opin.
detect: a new screening questionnaire to identify neu- 2019;35(7):1177–85. https://doi.org/10.1080/030079
ropathic components in patients with back pain. Curr 95.2018.1562687.
Med Res Opin. 2006;22(10):1911–20. https://doi.org/ 55. Rice ASC, Finnerup NB, Kemp HI, Currie GL,
10.1185/030079906X132488. Baron R.  Sensory profiling in animal models of
49. Attal N, Fermanian C, Fermanian J, Lanteri-Minet neuropathic pain: a call for back-translation. Pain.
M, Alchaar H, Bouhassira D.  Neuropathic pain: are 2018;159(5):819–24. https://doi.org/10.1097/j.
there distinct subtypes depending on the aetiology or pain.0000000000001138.
anatomical lesion? Pain. 2008;138(2):343–53. https:// 56. Sveen KA, et  al. Small- and large-fiber neuropathy
doi.org/10.1016/j.pain.2008.01.006. after 40 years of type 1 diabetes: associations with
50. Scholz J, et  al. A novel tool for the assessment glycemic control and advanced protein glycation:
of pain: validation in low back pain. PLoS Med. the Oslo Study. Diabetes Care. 2013;36(11):3712–7.
2009;6(4):e1000047. https://doi.org/10.1371/journal. https://doi.org/10.2337/dc13-­0788.
pmed.1000047. 57. Goyal SN, et  al. Challenges and issues with
51. Baron R, Tölle TR, Gockel U, Brosz M, Freynhagen streptozotocin-­induced diabetes  - a clinically rel-
R.  A cross-sectional cohort survey in 2100 patients evant animal model to understand the diabetes
with painful diabetic neuropathy and postherpetic pathogenesis and evaluate therapeutics. Chem Biol
neuralgia: differences in demographic data and sen- Interact. 2016;244:49–63. https://doi.org/10.1016/j.
sory symptoms. Pain. 2009;146(1):34–40. https://doi. cbi.2015.11.032.
org/10.1016/j.pain.2009.06.001. 58. Klinck MP, et  al. Translational pain assessment:
52. Bouhassira D, et  al. Development and validation could natural animal models be the missing link?
of the neuropathic pain symptom inventory. Pain. Pain. 2017;158(9):1633–46. https://doi.org/10.1097/j.
2004;108(3):248–57. https://doi.org/10.1016/j.pain. pain.0000000000000978.
2003.12.024. 59. Bouhassira D, et  al. Stratification of patients based
53. Spallone V, Greco C. Painful and painless diabetic neu- on the neuropathic pain symptom inventory: develop-
ropathy: one disease or two? Curr Diab Rep. 2013;13:7. ment and validation of a new algorithm. Pain. 2020.
https://doi.org/10.1007/s11892-­013-­0387-­7. https://doi.org/10.1097/j.pain.0000000000002130
Neurotrophic Factors
in the Pathogenesis and Treatment
of Diabetic Neuropathy

Nigel A. Calcutt

1 Introduction cells, satellite cells, macrophages, and neurons


themselves secrete multiple neurotrophic and
Neurotrophic factors may be broadly described other factors and express a veritable cornucopia
as polypeptides that signal through receptor tyro- of receptors (Fig. 1).
sine kinases to support survival, growth, and dif- Diabetic neuropathy is commonly described
ferentiation of developing neurons and/or as a slowly developing dying back axonopathy
maintain the phenotype of adult neurons. Nerve that presents as a distal symmetrical polyneurop-
growth factor (NGF), the prototypical neuro- athy [5]. The impact of neurotrophic deficiency
trophic factor, was initially studied as a critical on maintenance of neuronal phenotype, nerve
molecule for the survival, growth, and differenti- terminals, and axonal structure in adult rodents is
ation of small sensory neurons in the peripheral consistent with this pathology and invites consid-
nervous system (PNS). While not required for eration of loss of neurotrophic support as a patho-
survival of adult sensory neurons, NGF regulates genic mechanism contributing to diabetic
both the phenotype and regenerative capacity of neuropathy. There is also a school of thought that
adult peripheral sensory neurons. The initial a primary Schwannopathy co-exists with axo-
model developed from the study of NGF, ideal- nopathy in diabetic neuropathy and thus may also
ized a neurotrophic factor polypeptide produced contribute to neurotrophic deficiency given the
and secreted by end organs such as muscle or growing appreciation that Schwann cells provide
skin that was bound and internalized by receptors continuous metabolic and trophic support to their
at the axonal terminals and retrogradely trans- axons [6]. The neurotrophic axis, representing
ported to the cell body as a signaling endosome neurotrophic factor synthesis, secretion, signal-
to influence gene expression and neuronal pheno- ing, axonal transport, and gene regulatory mech-
type [1, 2]. However, this model has been repeat- anisms, is complex and offers multiple sites for
edly decorated by the ever increasing appreciation lesions that induce a loss of neurotrophic support.
of the complexity and subtlety of cell:cell inter- Delivery of exogenous factors to replace defi-
actions [3, 4]. Within the PNS, neurotrophic fac- cient endogenous trophic support is an obvious
tors may act as autocrine regulators as well as therapeutic approach, provided that any impair-
classical paracrine messengers, while Schwann ment of receptor mediated uptake or downstream
components of the neurotrophic axis in target
N. A. Calcutt (*) neurons can be overcome. Providing excess neu-
Department of Pathology, University of California rotrophic support may also allow neurons to
San Diego, La Jolla, CA, USA
e-mail: ncalcutt@ucsd.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 127
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_8
128 N. A. Calcutt

Fig. 1  Neurotrophic support in the PNS.  The original arrows) may also be supplied by cells of the spinal cord
model of end-organ derived factors (red arrows), such as (neurons, microglia, astrocytes, and oligodendrocytes)
muscle-derived BDNF (upper) and skin-derived NGF and peripheral nerve (Schwann cells, satellite cells, and
(lower), binding p75NTR/trk receptor complexes before resident macrophages), while neurons also export trophic
internalization and retrograde transport to the nucleus has factors to their peripheral and central terminals
been augmented by appreciation that factors (orange

resist the multiple other metabolic insults directed


at them by diabetes or promote recovery from the 2 The Neurotrophin Family
impact of such insults, irrespective of whether
impaired neurotrophic support is the primary All polypeptides in this family share about 50%
pathogenic lesion. sequence homology and signal through receptors
This review will cover the impact of diabetes that combine a common low affinity sub-unit
upon both the classic neurotrophic factor fami- (p75NTR) and a high affinity ligand-specific tyro-
lies and also other factors that have been exten- sine kinase sub-unit, the tropomyosin receptor
sively studied in the context of their disorder kinases A-C (Trk’s A-C). Neurotrophin precur-
potentially contributing to the pathogenesis of sors (pro-neurotrophins) may also be secreted
diabetic neuropathy and/or their potential as and signal through p75NTR that associates with
therapeutics to treat diabetic neuropathy. The other sub-units [10] while the p75NTR sub-unit
complexities of the impact of diabetes on the acting as a monomer transduces signals that pro-
contribution of neurotrophic factors to injury mote neuronal apoptosis and growth cone col-
responses such as nerve regeneration following lapse [11]. The emergence of the roles of NGF
physical lesion, inflammation, and wound heal- and other neurotrophins in neuronal development
ing in skin have been recently reviewed in detail and maintenance of the adult phenotype and
elsewhere [7–9] and are mentioned only en regeneration has been paralleled by interest in
passant. whether disrupted neurotrophic support
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 129

c­ ontributes to diabetic neuropathy and the viabil- ings encouraged clinical trials of NGF as a ther-
ity of neurotrophins as therapeutics. apy for diabetic neuropathy. Unfortunately, the
efficacy of injected recombinant human NGF
NGF  In the adult PNS, the TrkA receptor sub-­ suggested by phase II clinical studies was accom-
unit that confers NGF sensitivity is expressed by panied by dose limiting side effects, including
sympathetic and predominantly small diameter pain, which prompted changes in formulation
nociceptive sensory neurons [12] and cells adja- and dose that may have contributed to the failure
cent to nerve ending such as keratinocytes and of subsequent phase III study [36]. While this
muscle secrete NGF.  In contrast to embryonic failure largely discouraged pursuit of NGF injec-
neurons, NGF is not a required factor for survival tion therapy, it is currently allowed for use against
of adult peripheral sensory neurons in vitro [13] diabetic neuropathy and other neurological dis-
and disruption of NGF in adult rodents does not eases in China [37] and in a recent exploratory
induce marked neuronal loss. However, NGF trial recombinant human NGF was reported to
depletion does impact the phenotype of NGF-­ alleviate indices of erectile dysfunction [38].
sensitive adult neurons, for example, by reducing Alternative experimental approaches designed to
expression of the neuropeptide neurotransmitters avoid NGF-induced injection site pain include
substance P and CGRP [14] and by reducing inducing site-specific expression of NGF via a
nociceptor sensitivity [15], thereby impeding viral vector [39] and inducers of local NGF pro-
heat pain perception [16]. Conversely, NGF also duction that concurrently improve indices of neu-
activates inflammatory cells and mediators, sen- ropathy in diabetic rodents. Examples range from
sitizes nociceptors, and induces both myalgia and botanical extracts [40] and vitamin derivatives
chronic injection site pain when given to healthy [41] to small molecules [42, 43], a CB-1 receptor
volunteers [17–19]. antagonist [44] and a PDE5 inhibitor [45].
However, these molecules also induce a complex
Expression of NGF and its receptor sub-units array of alternative potentially neuroprotective
is reduced in skin, and muscle of diabetic rodents responses and ascribing efficacy specifically to
and the retrograde axonal transport of locally induction of NGF has not been possible. As dis-
produced or injected NGF is also reduced [20– cussed below, the ability to modify NGF to
23]. This diminution of NGF support is consis- exclude its algogenic capacity and allow pain
tent with the parallel diabetes-induced reduction free delivery has recently emerged as a novel
in protein levels and axonal transport of NGF-­ therapeutic approach [46].
regulated gene products such as substance P and The suggestion that NGF is involved in gener-
CGRP [20, 24–28] that, in turn, leads to sensory ating some forms of chronic pain has led to anti-­
dysfunction via reduced peripheral and spinal NGF therapies being explored as an approach to
release of these neuropeptides during stimulation suppressing pain states [47, 48]. For example, the
of sensory neurons [29–33]. The capacity of NGF recombinant humanized monoclonal antibody
to both promote sensory nerve growth and regu- Tanezumab, which binds NGF and prevents
late nociceptor phenotype has prompted investi- interaction with its receptor, was developed for
gation of its potential as a therapy for diabetic use against osteoarthritis [49] and chronic low
neuropathy. back pain [50, 51] despite initial concerns regard-
Multiple indices of small fiber neuropathy in ing potential accelerated damage to joints that
diabetic rodents are prevented or reversed by sys- disrupted development programs [52].
temic or intrathecal injection of NGF [22, 26, Tanezumab also produced clinically significant
34]. There are also reports of an impact on indi- improvement of pain scores above placebo lev-
ces of large fiber neuropathy [35], although this is els, without worsening neuropathy, in a small
presumably an indirect effect given the absence cohort of patients with painful diabetic neuropa-
of TrkA receptor expression by most large thy [53] and a more expansive phase 2 clinical
myelinated fibers. These positive preclinical find- trial has recently reported data (NCT01087203).
130 N. A. Calcutt

BDNF  BDNF is widely studied because of its is consistent with the absence of TrkB on most
roles in the brain in relation to development and small sensory fibers. It also had no effect on
modulation of synaptic activity and plasticity. In conduction deficits and myelin damage in large
the context of diabetes, reduced BDNF expres- sensory fibers of galactose-fed rats (a model of
sion is implicated in encephalopathy [54], polyol pathway hyperactivity) but did protect
dementia [55], and retinopathy [56]. A variety of against these disorders in large motor fibers
cells associated with the PNS, including muscle, [64]. However, a subsequent clinical trial of
keratinocytes, and Schwann cells express BDNF recombinant human BDNF in diabetic subjects
and induction of BDNF following nerve injury with neuropathy failed to demonstrate improve-
plays a role in neuronal survival and/or regenera- ment of large fiber conduction slowing after 3
tion [57]. Responsiveness to BDNF is conferred months of treatment in a small group of sub-
by expression of the TrkB receptor sub-unit jects, although improvement in cool thermal
operating in conjunction with the p75NTR. TrkB detection thresholds was noted [73]. As with
is localized to 10–25% of sensory neurons in the NGF (see above), BDNF is induced by a variety
dorsal root ganglia, mainly large myelinated of potential therapeutic agents and has been
fibers, and to motor neurons, Schwann cells and linked to their efficacy against indices of neu-
post-synaptic muscle at the neuromuscular junc- ropathy in diabetic rodents, although multiple
tion [58]. Small, NGF-sensitive sensory neurons other plausible therapeutic pathways are also
also constitutively express BDNF and transport modulated and causality has not be established
it to their central terminals in large dense core [66, 67, 74, 75]. BDNF mimetics have been
(peptidergic) vesicles where, upon release, it developed and promote nerve growth [76, 77]
modulates nociceptive input by enhancing but efficacy against diabetic neuropathy has yet
NMDA receptor-mediated glutamatergic excit- to be reported.
atory signaling [59–62]. Thus, broadly, in the
PNS, motor and some large sensory fibers are BDNF and Pain  A role for BDNF in spinally-­
responsive to BDNF while a proportion of small mediated pain during diabetes is suggested by
sensory neurons produce BDNF and secrete it studies showing that indices of painful neuropa-
centrally to modulate nociception (Fig. 1). thy in diabetic rats can be suppressed by intrathe-
cal delivery of a TrkB/Fc chimera acting as a
Hyperglycemic rodents show increased BDNF decoy [78, 79]. The source of spinal
BDNF mRNA and protein in muscle, which has BDNF in diabetes has been attributed to activated
been proposed to reflect a response to metabolic microglia [79], glial cells that are increasingly
injury [21, 63–65]. In contrast, whole nerve described as modulators of nociceptive input via
BDNF mRNA and protein levels are reduced release of a variety of inflammatory and pro-­
[66–68], possibly a consequence of nociceptive factors, including BDNF, that impact
hyperglycemia-­induced impaired expression by both pre- and post-synaptic neurons [80–82].
Schwann cells [69]. Diabetes also impedes Primary afferents represent another potential
peripherally-derived support of large fibers by source of increased BDNF release in the spinal
BDNF as there is a decrease in TrkB receptor cord [83]. BDNF expression increases dramati-
mRNA in sciatic nerve [70] and reduced retro- cally after inflammatory or physical injury,
grade axonal transport of endogenous BDNF including induction in larger sensory neurons
from the periphery [71]. These observations led that mediate allodynia to light touch [84, 85].
to studies investigating the efficacy of BDNF While there is reduced expression of BDNF by
replacement therapy against indices of neuropa- Schwann cells in the sciatic nerve and spinal
thy in diabetic rodents. BDNF injections did not roots in diabetic rodents, diabetes increases
prevent indices of small fiber dysfunction, such BDNF mRNA and protein in DRG [21, 74, 86,
as reduced neuropeptide expression [26] and 87] suggesting the potential for increased central
thermal hypoalgesia [72] in diabetic rats, which transport and release.
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 131

Mechanisms by which BDNF promotes tion, and a state of spinal disinhibition that drives
spinally-­mediated pain in diabetes may include or amplifies pain-associated behaviors [91].
increased NMDA receptor signaling in neurons KCC2 expression is reduced in diabetic rats with
and microglia [88] and disruption of GABAergic an attendant shift of spinal GABAergic function
inhibitory tone. The latter is a consequence of the from inhibitory to excitatory, so that spinal deliv-
capacity of BDNF to regulate expression of the ery of GABAA antagonists blocks indices of pain
membrane ion pump, potassium/chloride co-­ rather than promoting it [92, 93]. Conversely,
transporter KCC2 [89]. During neuronal develop- incapacitating spinal BDNF in diabetic rats
ment, BDNF levels are high, KCC2 pump restores KCC2 expression, GABAergic inhibitory
expression and activity is low and the post-­ function and normalizes pain behavior [78, 79].
synaptic membrane chloride gradient is such that Whether there is disruption of the spinal
activation of the GABAA receptor (a chloride BDNF/KCC2/GABAA receptor axis in humans
channel) is excitatory. Upon maturation of the with painful diabetic neuropathy is not known.
nervous system, decreased expression of BDNF However, the phenomenon of rate-dependent
results in increased expression of KCC2 which depression (RDD) of the electromyogram H
increases the membrane chloride equilibrium wave following repeated stimulation of periph-
potential and switches GABAA receptor function eral nerve also reflects spinal GABAergic inhibi-
to inhibitory [90]. Elevated spinal BDNF follow- tory function in normal animals and there is loss
ing nerve injury in adult animals has been linked of RDD in diabetic rodents [78, 92, 94–96]. Loss
to decreased KCC2, loss of GABAergic inhibi- of RDD (Fig. 2) has been proposed as an electro-

Fig. 2  BDNF-mediated spinal disinhibition. Diabetes-­ ing to increased pain perception (purple arrows) and loss
induced increased BDNF released from primary afferents of rate-dependent depression of the H wave (RDD) in the
and/or activated spinal microglia reduces spinal KCC2 EMG following repetitive peripheral stimuli (blue
expression, which results in the GABAA receptor losing arrows). Loss of RDD may therefore serve as a clinical
inhibitory function and possibly assuming an excitatory biomarker for patients with painful diabetic neuropathy
function. Loss of spinal GABAergic inhibition has two driven by reduced spinal disinhibition
consequences: disinhibition of sensory throughput lead-
132 N. A. Calcutt

physiological biomarker of spinally-mediated port of NT-3 to the neuronal perikaryon and


pain arising from an abnormal BDNF/KCC2/ sub-optimal neurotrophic support [70, 108, 109].
GABA axis [78, 86]. Consistent with this hypoth- Similar to BDNF (see above), impaired target-­
esis, cohorts of subjects with type 1 or type 2 dia-derived support is accompanied by increased
betes with painful diabetic neuropathy showed expression of NT-3 mRNA in dorsal root ganglia,
impaired RDD compared to non-diabetic sub- which may reflect a compensatory mechanism
jects or patients with painless diabetic neuropa- [110]. In contrast, both TrkC mRNA and NT-3
thy [97, 98]. As only a sub-group of the painful protein are increased in the skin of diabetic sub-
diabetic neuropathy group showed markedly jects with neuropathy and it has been suggested
abnormal RDD, this disorder has the potential to that this reflects a response to ongoing denerva-
serve as a biomarker for identifying patients in tion [111, 112].
whom pain is mediated primarily by altered spi- The capacity of NT-3 to support a broader
nal disinhibition [86, 97]. Interestingly, a sub-­ array of peripheral neuron types than other neu-
group of patients with painless neuropathy rotrophins makes it an attractive potential ther-
showed exaggerated RDD, raising the possibility apeutic to treat diabetic neuropathy, albeit with
of enhanced spinal GABAergic inhibition con- the potential caveat of having to negotiate
tributing to an absence of pain [98]. reduced TrkC expression by target neurons
[108]. Systemic delivery of NT-3, protected
NT-3  Within the PNS, the TrkC receptor sub-­ large fiber conduction velocity in rat models of
unit that confers NT-3 selectivity is found in hyperglycemia [113, 114] and diminished axo-
motor neurons, large diameter sensory neurons nal transport of diabetes-induced stress signals
innervating muscle, and sympathetic neurons [115] whereas it was without effect on indices
[99, 100]. Low affinity binding to combinations of autonomic neuropathy [116]. Exercise-
of p75NTR and Trk’s A or B also occurs [101] induced induction of NT-3 expression in the
making NT-3 the most promiscuous of the neuro- muscle of diabetic rats is also accompanied by
trophins with potential to provide the broadest prevention of nerve conduction slowing,
therapeutic coverage. NT-3 itself is expressed by although causality was not confirmed [117].
skeletal muscle and retrogradely transported in Delivery of NT-3 to central terminals of pri-
the manner of a classical target-derived trophic mary afferents via intrathecal injection pre-
factor [102]. However, there is also evidence that vented loss of dermal myelinated fibers and
NT-3 undergoes anterograde axonal transport in increased Merkel cell number in diabetic mice;
certain neuronal populations and, as discussed but did not impact mechanical hypoalgesia
above for BDNF, may serve as a modulator of [118]. Interestingly, when NT-3 therapy was
synaptic transmission [103, 104] and synaptic combined with NGF it diminished the efficacy
organization [105] in the spinal cord. TrkC is of NGF alone, possibly by competing with
expressed in the corticospinal tract, fasciculus NGF for the p75NTR [118]. The mechanisms by
cuneatis and fasciculus gracilis of the spinal cord which NT-3 protects neurons from diabetes-­
and NT-3 has been explored to treat spinal cord induced damage include normalization of cal-
injury [106]. cium homeostasis [119], prevention of
mitochondrial dysfunction [120], and mainte-
Reduced NT-3 has been reported in the muscle nance of appropriate neurofilament organiza-
of diabetic patients, especially those with neu- tion [121].
ropathy and correlated with muscle weakness NT-4, like BDNF, signals through the TrkB
[107]. Expression of NT-3 mRNA and protein is receptor but studies of this neurotrophin are lim-
also reduced in the skeletal muscle of type 1 dia- ited to a report of decreased NT-4 in the sciatic
betic rodents and, in combination with reduced nerve of diabetic rats [70], while in human ­muscle
neuronal expression of the TrkC receptor sub-­ NT-4 expression parallels the decrease seen in
unit, results in a reduced retrograde axonal trans- NT-3 [107].
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 133

3 GDNF Family Ligands lished by delivery of the factor direct to the spinal
cord of diabetic mice that demonstrated efficacy
Members of the GDNF family ligands: glial cell against loss of non-peptidergic neuronal termi-
line-derived neurotrophic factor (GDNF), arte- nals in the spinal cord [136] and skin [133] and
min, neurturin, and persephin, exhibit relatively also loss of responses to chemical stimuli [137].
low amino acid sequence homology but are More recently, small molecule RET agonists
placed in the same family based on their activa- [138] or modulators of RET signaling [139] have
tion of the tyrosine kinase receptor “rearranged shown efficacy against behavioral indices of sen-
during transfection” (RET). RET is activated fol- sory dysfunction in diabetic rodents. Of other
lowing binding of a factor to the co-receptor, members of the family, neurturin also protected
GDNF receptor alpha (GRFα), which itself has against loss of cutaneous nerves [133].
four variants (GRFα 1-4) that confer ligand selec-
tivity or specificity [122]. Activation of RET trig-
gers signaling through one or more multiple 4 IL-6 Cytokine Family
cascades, including the MAPK, PI3K, Erk, and
Akt pathways, depending on the specific ligand/ This superfamily of cytokines is defined by their
receptor configuration and cell. In Schwann cells signaling through a receptor complex that shares
and select neuronal populations, GDNF can also a common gp130 sub-unit that interacts with
signal through GRFα in the absence of RET cytokine-specific sub-units. It includes
[123] while interaction of activated RET with the Interleukins 6, 11, 27, and 30, along with ciliary
neurotrophin p75NTR sub-unit has also been neurotrophic factor (CNTF), leukemia inhibitory
described as part of what is increasingly viewed factor (LIF), oncostatin M (OSM), neuropoietin,
as a complex mechanism for cross-talk between a cardiotrophin-like cytokine factor (CLCF1), and
variety of neurotrophic factors [124]. cardiotrophin-1 (CT-1). A sub-group, comprising
GDNF can be viewed as a classic target CNTF, LIF, OSM, CLCF1, and CT-1 share both
derived neurotrophic factor for motor neurons gp130 and the LIF receptor (LIFR) sub-units and
(Fig. 1) as it is expressed by adult skeletal mus- signal via the JAK/STAT, MAPK, and PI3K/Akt
cle, binds GRFα1/RET located on motor neuron pathways so that there is some overlap in their
synaptic terminals, is internalized and then retro- phenotypic consequences, such as capacity to
gradely transported and promotes axonal growth, regulate metabolism and impact body weight
synapse formation, and modulation of pre- and [140, 141]. While the ability to reduce food con-
post-synaptic function [125, 126]. GFR/RET sig- sumption, blood glucose and body weight in
naling also occurs in sub-populations of primary obese models of type 2 diabetes [142] may have
sensory neurons and postganglionic autonomic indirect impact on the nervous system and dia-
neurons during development [127–129]. In adult betic neuropathy, there is also evidence for direct
DRG around 35% of neurons, predominantly actions of some members of this group of cyto-
small non-peptidergic neurons that label with kines on the PNS.
IB4 and lack the TrkA receptor sub-unit, are CNTF is a 22-kDa polypeptide neurotrophic
GDNF responsive [130] while neurons and factor that is expressed in a wide range of tissues
Schwann cells increase expression of GDNF fol- including bone marrow, testis, and heart, but
lowing injury [131, 132]. which has been widely studied to date in the con-
Diabetic rodents [133] and humans [134] have text of actions in the nervous system. Cells
been reported to exhibit increased mRNA for responsive to CNTF express the glycosyl-­
GDNF in skin, whereas GDNF mRNA and pro- phosphatidylinositol (GPI) linked CNTF
tein are decreased in nerve from diabetic rats ­receptor sub-unit [143] which confers high affin-
[135], suggesting a complex progression of ity CNTF binding and signaling activity when
diabetes-­induced lesions and responses to injury. combined with both the LIFR and the gp130
The therapeutic potential of GDNF was estab- sub-­units. CNTF also shows low affinity binding
134 N. A. Calcutt

to the IL-6R [144]. Early studies showed CNTF glycemia driven excess polyol pathway flux
expression localized to Schwann cells of periph- may contribute to the pathogenesis of diabetic
eral nerve [145, 146] with levels increasing dur- neuropathy by disrupting expansion of axonal
ing post-natal development to steady state in diameter (and hence conduction velocity) sec-
adult nerve. Nerve lesion results in extracellular ondary to depletion of Schwann cell derived
accumulation of CNTF at the lesion site fol- CNTF.  How CNTF interacts with axons
lowed by downregulation of the factor as remains unclear given the absence of an estab-
Schwann cells enter their non-myelinating pro- lished secretory mechanism, although direct
liferative stage and then a restoration of expres- transfer between Schwann cell and axonal
sion that accompanies re-myelination [147–149]. cytoplasm is plausible given that other cyto-
The ability of CNTF to prevent neuronal death plasmic constituents undergo such transfer
and promote axonal growth after lesion [150– [162–169].
152], along with the absence of a known secre- The therapeutic potential of CNTF was
tion mechanism, prompted the suggestion that determined by treatment with recombinant
CNTF release from damaged Schwann cells pro- CNTF, which prevented impaired nerve regen-
vides an immediate neuroprotective response to eration after sciatic nerve crush in diabetic rats
physical nerve injury [152, 153] that precedes [170]. Impaired corneal nerve regeneration fol-
the temporally-regulated increase in production lowing physical lesion in diabetic mice and the
of other growth factors by Schwann cells and degeneration of amacrine and retinal ganglion
other cells at the lesion site. This has led to many cells in diabetic rats were also prevented by
studies that have looked to exploit local delivery local CNTF delivery [171, 172]. These findings
of CNTF and other growth factors to promote are consistent with the capacity of CNTF to
peripheral nerve regeneration after traumatic promote axonal regeneration, including
injury [154, 155]. Aside from a role following enhancement of neurite outgrowth from sen-
traumatic injury, CNTF also appears to contrib- sory neurons derived from diabetic rodents
ute to normal maintenance of peripheral motor [173]. Exogenous CNTF also ameliorated paw
neurons. The PNS of CNTF-­null mice develops thermal hypoalgesia, reduced axonal caliber
normally but there is loss of facial motor neurons and the accompanying motor and sensory nerve
during adult life [156] and morphological abnor- conduction slowing of uninjured nerve in dia-
malities of the paranode and myelin in motor betic rodents [72, 170, 173]. These effects were
neurons of L5-L6 spinal ventral roots between 3 seen in the absence of impact of CNTF on
and 9 months of age accompanied by impaired hyperglycemia and suggest neuroprotective
axonal diameter maturation [157]. Depletion of actions independent of nerve injury and repair
Schwann cell derived CNTF therefore has the programs. Putative mechanisms that may
capacity to contribute to the pathogenesis of impact both nerve repair after physical injury
both axonal and glial manifestations of periph- and resistance to metabolic insult involve resto-
eral neuropathy. ration of normal mitochondrial function in the
Rat models of diabetic neuropathy develop nerve of diabetic rodents via activation of JAK/
reduced CNTF bioactivity and protein levels in STAT signaling and NF-KB [173, 174].
peripheral nerve over a time period that encom- Unfortunately, early clinical trials of systemic
passes onset of early physiological and struc- CNTF in subjects with motor neuron disease
tural indices of neuropathy, such as NCV identified a variety of dose-related and dose
slowing and reduced axonal caliber [158–160]. limiting side effects ranging from cough to
CNTF depletion and the accompanying reduc- anorexia and weight loss [175] that largely dis-
tion of axonal caliber were prevented by aldose couraged further development of the factor as a
reductase inhibition [159] a therapeutic therapy, although potential use as a weight loss
approach that targets metabolism of excess agent was subsequently explored [176]. As with
glucose in Schwann cells [161]. Thus, hyper- many neurotrophic factors, systemic delivery of
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 135

CNTF is unlikely to be viable due the diverse sub-units in sciatic nerve after onset of diabe-
array of cells and tissues that express the CNTF tes has been reported [191] while attenuated
receptor and the many biological pathways it induction of LIF and IL-6 in sensory and auto-
activates. Localized delivery to treat degenera- nomic ganglia with reduced STAT signaling
tive eye diseases has shown promise [177, 178] and reduced expression of regeneration associ-
and, as discussed below, use of engineered cells ated genes is seen following nerve crush injury
that distribute to specific parts of the body and to diabetic mice and accompanies impaired
secrete CNTF as part of a cocktail of supportive nerve regeneration [192]. However, as yet
factors [179] may offer the most practical these observations have not been exploited to
approach for any future use against diabetic investigate new therapeutic approaches for dia-
neuropathy. betic neuropathy.
Of other members of this cytokine family,
the IL-6 receptor sub-unit (IL-6R) is expressed
by myelinating Schwann cells and localizes to 5 Insulin, C-Peptide,
the nodes of Ranvier [180]. There is also a sol- and Insulin-Like Growth
uble form of the receptor sub-unit (sIL-6R) Factors
that binds secreted IL-6 and can interact with
membrane bound gp130 to confer IL-6 sensi- Unlike muscle and fat, where glucose uptake
tivity on cells that do not themselves express occurs via the insulin-regulated GLUT4 trans-
IL-6R, in a process termed trans-signaling porter, perineurial and endothelial cells of the
[181]. Following physical injury, IL-6 is blood:nerve barrier utilize the insulin-­
secreted by denervated Schwann cells and independent GLUT1 glucose transporter [193]
IL-6R expression increases suggesting roles while both GLUT1 the insulin-independent
for IL-6 in axon:Schwann cell interactions dur- GLUT3 high affinity glucose transporter are
ing maintenance of steady state and for myelin expressed by axons and Schwann cells [194].
clearance via monocyte recruitment and re- Consequently, nerve glucose levels directly
myelination during nerve regeneration [182– reflect plasma glucose levels during hyperglyce-
184]. There are reports that IL-6 treatment mia. Agents that block metabolism of excess glu-
ameliorated multiple indices of neuropathy in a cose in nerve without impacting systemic insulin
rat model of type 1 diabetes [185–187] and or glucose levels, such as aldose reductase inhibi-
suggestions that it may be a viable therapy tors, prevent and reverse multiple indices of neu-
under controlled release conditions [188]. ropathy in diabetic rodents [195], although to
However, it is becoming increasingly appreci- date they have failed to translate to clinical use
ated that IL-6 has differing properties depend- [196]. This glucotoxicity, along with the occur-
ing on whether it signals via membrane bound rence of peripheral neuropathy in both type 1 and
(classical signaling, broadly anti-­inflammatory) type 2 diabetes, was historically cited as a ratio-
or soluble (trans-signaling, broadly pro-inflam- nale for hyperglycemia being the primary patho-
matory) IL-6R [181]. Recent interest in the genic mechanism that initiated diabetic
role of IL-6 in diabetic neuropathy has largely neuropathy [197]. However, both secreted prod-
shifted to the pro-inflammatory actions of IL-6 ucts of pro-insulin cleavage, insulin and
and its potential role in diabetic neuropathic C-peptide, have neurotrophic properties and it is
pain that have arisen from reports of increased increasingly recognized that insulinopenia may
expression of IL-6 in peripheral nerve and spi- produce a deficit in trophic support to neurons
nal cord [189, 190]. that contributes to the pathogenesis of diabetic
There have been few studies of other mem- neuropathy. Impaired insulin signaling also
bers of the IL-6 cytokine family in the context occurs in the brain during diabetes [198, 199] and
of diabetic neuropathy. A transient increase contributes to cognitive impairments associated
expression of the gp130 and LIFR receptor with both diabetes and also other neurodegenera-
136 N. A. Calcutt

tive diseases such as Alzheimer’s Disease. The type 1 diabetic rodents where insulin was pro-
role of insulin deficiency in CNS pathology, in vided at levels that did not impact hyperglyce-
terms of both its metabolic and neurotrophic mia. Low doses of systemically-­ delivered
effects has been extensively reviewed insulin correct impaired calcium homeostasis in
[200–202]. the perikarya of sensory neurons from diabetic
rats [119] and protect neuronal mitochondrial
Insulin  High affinity insulin receptors are pres- phenotype and function [119, 207, 218–220].
ent on endothelial cells, pericytes, Schwann cell, This is accompanied by prevention or attenua-
and neurons in the PNS [203–205]. In both tion of multiple indices of large and small fiber
Schwann cells and axons, insulin signaling neuropathy. Similar efficacy against functional
drives growth and maturation via the phos- and structural markers of neuropathy is seen
phoinositide 3-kinase (PI 3-kinase)/Akt signal- when low dose insulin is delivered by intrana-
ing pathway [206, 207]. The direct effect of sal, intrathecal or intraneuronal injection [204,
insulin in promoting axonal growth was first 221–225].
demonstrated using sensory neurons derived
from adult rodents [208]. While embryonic C peptide  C peptide is a 31 amino acid poly-
knockout of neuronal insulin receptors did not peptide that connects the A and B chains of
generate a peripheral neuropathy phenotype insulin within the pro-insulin molecule, sepa-
[209], dual knockout of receptors for insulin, rated from the insulin chains prior to secretion
and the related factor IGF-1  in Schwann cells and released from pancreatic Beta cells in equal
resulted in the development of peripheral nerves amounts to insulin. While initially considered
exhibiting both reduced axonal diameter and inert, it has been argued that C-peptide has a
thin myelin that persisted into adulthood. Adult physiologically relevant role in modulating
mice also displayed multiple indices of both insulin signaling and that C-peptide deficiency
large and small fiber neuropathy that replicated contributes to the disease phenotype of type 1
those seen in diabetic mice [206]. Insulin may diabetes [226, 227], including the more severe
therefore be viewed as trophic factor for periph- neuropathy reported in type 1 versus type 2 dia-
eral nerve and insulin deficiency or receptor dys- betic rats [212]. C-peptide promotes intracellu-
function has the potential to contribute to lar signals in multiple cell types, including
neuropathy independent of glycemic status [210, peripheral nerves [228] and the orphan G pro-
211]. tein coupled receptor GRP146 has been pro-
posed as one of potentially many C peptide
Studies directly comparing rat models of receptors [229], although concerns of these data
type-1 and type-2 diabetes with equivalent remain [230].
hyperglycemia demonstrated that indices of
neuropathy develop earlier, and are more Treating diabetic rodents with C-peptide
extreme, in the insulin-deficient animals, sup- prevents or reverses multiple indices of neu-
porting the argument that insulin deficiency ropathy with a mechanism of action that
contributes to neuropathy in type-1 diabetes includes potentiation of insulin signal transduc-
[212, 213]. There is also a growing suspicion tion, induction of other neurotrophic factors,
that neuropathy phenotype, and indeed patho- and stimulation of nitric oxide-mediated vaso-
genesis, may vary between type 1 and type 2 dilation [231, 232]. Similar efficacy was dem-
diabetes [214, 215], although both forms of the onstrated for a long acting analog of C-peptide
disease incorporate impaired insulin signaling [233] and this molecule was advanced to phase
in rodent nerve [216, 217]. Further evidence for III clinical trial in patients with type 1 diabetes
a role of impaired insulin-­mediated trophic sup- following promising outcomes when using
port in neuropathy comes from animal studies in native C-peptide in exploratory studies [234–
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 137

236]. Long acting C-peptide significantly persistent feature in insulin-deficient diabetic rats
improved sensory nerve conduction from base- [243–245]. Nerve IGF-2 is also reduced in rat
line but a similar effect was seen in the placebo models of type-2 diabetes and, while plasma
group so that the primary efficacy measure was IGF-1 and nerve IGF-1 mRNA remain normal
not supported [237]. C-peptide also signifi- [246, 247], nerve of type 2 diabetic mice shows
cantly improved vibration perception threshold IGF-1 resistance that parallels insulin resistance
against both baseline and placebo but this posi- [216]. The complex interactions of IGF, IGFBP,
tive effect on a patient reported outcome was and IGFR have also been investigated [248, 249].
not considered sufficient to advance the thera- It has been suggested that downregulation of
peutic approach further. IGF-1 in Schwann cells is secondary to increased
glucose metabolism through the polyol pathway
Insulin-like Growth Factors  IGF-1 and IGF-2 [250]. Given the broad impact of IGFs on periph-
are single chain polypeptides that share around eral nerve, such findings prompted multiple pre-
45% sequence homology to pro-insulin. They clinical studies to assess efficacy of systemic or
have a variety of survival and growth related intrathecal delivery of IGF-1 in diabetic rodents
effects throughout the body, with IGF-2 being and there are many reports of improved func-
particularly important during development, tional and structural indices of neuropathy [79,
while the effects of IGF-1 extend throughout 204, 225, 247, 251–257]. A recent study found
life [238]. Both IGFs bind to the IGF-1 receptor that IGF-1 causes acute injection site hyperalge-
(IGF-1R) which signals through tyrosine sia in diabetic mice [258] so reports that local
kinases and multiple downstream pathways IGF-1 and/or IGF 1R expression may be induced
including the PI 3-kinase/Akt and Ras/Erk cas- by a variety of agents that concurrently impact
cades. IGF-1 also binds to the insulin receptor neuropathy in diabetic rodents [259–261] may
and insulin can activate IGF-1R so, while each provide an alternative to direct injection of the
factor has higher affinity for its own cognate factor.
receptor, there is a degree of functional overlap The pertinence of IGF replacement therapy in
[239]. A second receptor, IGF-2R selectively humans is supported by reports that circulating
binds IGF-2 but does not transduce a signal, IGF-1 is reduced in both type 1 and type 2 dia-
serving instead to reduce circulating IGF-2 by betic patients [262, 263] and that the deficit is
sequestering, internalizing, and degrading it. more extreme in those with neuropathy [264].
There is further regulation of circulating IGF’s However, the subtleties of the IGF/IGFBP/IGFR
by six IGF binding proteins (IGFBP1-6) and axis remain largely unexplored and the pleiotro-
assorted proteases that regulate the IGFBP’s. pic systemic effects of IGFs have caused their
The IGF-1R is expressed throughout the periph- therapeutic development to proceed with caution
eral nervous system and IGF-1 has effects on in recognition of the potential for multiple side
Schwann cells and sensory, motor, and auto- effects. Another caveat to the development of
nomic neurons. Mice deficient in IGF-1, or IGF’s as potential therapeutics is the need to con-
IGF-1R or that overexpress IGFBP5 show func- sider the impact of the IGFBP, which modulate
tional and structural pathology [240] indicating the bioavailability of circulating IGFs. Circulating
a role in maturation and maintenance of the levels of IGFBP1 are increased in type-1 diabetic
PNS, while IGF-1 also plays a prominent role in patients with neuropathy [265], while nerve from
orchestrating many aspects of nerve regenera- a small group of diabetic subjects with neuropa-
tion after injury [241]. thy showed reduced mRNA for IGF-1 but dra-
matically increased expression of IGFBP5
Reduced plasma levels of IGF-1 [242] and protein with the latter proposed to sequester
reduced expression of mRNA for IGF-1, IGF-2, IGF-1 and thereby reduce it effective concentra-
and IGF-1R by peripheral nerve are an early and tions in neurons [266].
138 N. A. Calcutt

There are multiple potential mechanisms by The hedgehog family of morphogens, com-
which IGFs may impact diabetic neuropathy prising sonic (SHH), desert (DHH), and Indian
given their pleiotropic metabolic and neuro- (IHH) regulate many aspects of embryonic
trophic capacity. For example, efficacy of sys- development including growth and patterning of
temic IGF-1 treatment may be secondary to the PNS [277]. They signal via a complex pro-
indirect effects, such as the induction of the cess that begins with binding to the transmem-
angiogenic and neurotrophic polypeptide vas- brane receptor patched that releases tonic
cular endothelial growth factor (VEGF) in inhibition of an associated transmembrane sig-
muscle and nerve of diabetic rodents [267, naling protein, smoothened (SMO). Disinhibition
268]. Direct effects of IGF-1 include protec- of SMO triggers signaling that activates multiple
tion of mitochondrial function [269] and aug- transcription factors to regulate cell phenotype.
mentation of ­ glycolytic capacity in neurons Consistent with their regulation of neuronal
[270], and regulation of Schwann cell differen- development, traumatic injury to the adult PNS
tiation status [250]. induces expression of both DHH and SHH, a
response that has been implicated in multiple
aspects of the nerve survival, repair, and regen-
6 Morphogens and Axonal eration programs [278]. A role in the develop-
Guidance Factors ment of endoneurial blood vessels has also been
suggested by studies in DHH knockout mice
Morphogens are key regulators of developmental [279]. DHH is expressed by Schwann cells in
biology that exert positive or negative influences adult peripheral nerve and mRNA levels are
on the patterns of tissue development by virtue of reduced by diabetes [279, 280], perhaps reflect-
their non-uniform distribution and consequent ing the metabolic damage to these cells that also
establishment of a concentration gradient. In the results in parallel CNTF depletion (see above).
context of development of the nervous system Treating diabetic rats with a SHH-­IgG fusion
there are many molecules that provide guidance protein ameliorated multiple indices of small
cues for axonal pathfinding including the ephrin, fiber neuropathy including the depletion of NGF
semaphorin, slit, and netrin families. Of these, protein and its retrograde axonal transport,
netrin has been reported to correct NCV slowing reduced neuropeptide proteins and reduced large
in adult diabetic mice via a presumed pro-­ fiber conduction velocity and axonal caliber
angiogenic mechanism [271]. Non-conventional [280]. Various small molecule agonists and
axonal guidance molecules include morphogens antagonists of the hedgehog/patched/Smo sig-
of the TGF-β, Wnt ligand, and hedgehog families naling pathways have been produced [281] and
[272]. In the adult PNS, TGF-β is expressed by an agonist of Smo, used to replicate the down-
neurons, Schwann cells, and pericytes and both stream consequences of hedgehog binding to
disrupts the basement membrane of the patched, prevented heat hypoalgesia and tactile
blood:nerve barrier and reduces neurite out- allodynia in diabetic mice [279]. My own expe-
growth in vitro [273, 274]. Expression of TGF-β rience is that efficacy of potent hedgehog path-
mRNA and protein is induced in the DRG and way agonists against indices of neuropathy in
nerve by diabetes, suggesting potential indirect diabetic rodents is offset by unwanted effects
and direct neurotoxic roles [273]. Similarly, it elsewhere (Fig. 3), illustrating the difficulties of
was recently reported that markers of Wnt signal- targeting growth factor therapies to nerve when
ing are increased in sciatic nerve, DRG, and spi- they have more catholic functions. The physical
nal cord of diabetic rats and that Wnt signaling effects illustrated in Fig.  3 are consistent with
inhibitors improved multiple behavioral, func- the role of exaggerated hedgehog signaling in
tional, and structural indices of neuropathy [275, basal cell carcinomas and the use of smo antago-
276]. nists to treat this condition [282].
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 139

MNCV

a 70 p<0.001 p<0.001 c

60
m/s

50

40
ic

st
l
ro

et

ni
nt

go
ia
co

+A
D

t ic
be
ia
D

Fig. 3 Hedgehog pathway agonism in diabetes. “fluff” (compare treated diabetic rat at left of panel b vs.
Streptozotocin-diabetic rats were treated with vehicle or a adjacent untreated diabetic rat) while the ventral skin
potent hedgehog signaling pathway agonist for 8 weeks. hypertrophied and depilated (panel c). After 7–8 weeks of
Agonist treatment prevented MNCV slowing (panel a). treatment rats began self-mutilating their ears and paws
However, agonist treatment also caused the dorsal fur to and were euthanized. Calcutt, unpublished

7 VEGF thelial growth factor (VEGF) also falls into this


category, as while largely studied as a vasculo-
For many years, loss of trophic support and genic and angiogenic factor that has potential to
nerve hypoxia secondary to microvascular dis- repair microvascular disease in diabetes, it also
ease have served as two parallel, often compet- has growth promoting properties when applied
ing, hypotheses to explain the pathogenesis of to neurons derived from adult rodents [285,
diabetic neuropathy. More enlightened views 286].
have considered them as two of many ongoing VEGFs form a sub-family of the platelet
stressors that cumulatively weaken the capacity derived growth factor (PDGF) family that con-
of neurons to sustain their most vulnerable and sists of PDGF’s A-D, VEGF’s A-D, and placenta
energy demanding terminal structures and growth factor (PGF). VEGF-A was the first
make nerves less able to survive and recover member of the VEGF sub-family to be described
from acute lesions. Angiogenic factors such as and exists in multiple isoforms due to alterna-
basic fibroblast growth factor (bFGF) show tive splicing of the VEGFA gene. All VEGFs
some efficacy against neuropathy and impaired bind VEGF receptors, of which there are at least
regeneration in diabetic rats [283, 284] while three sub-types (VEGFR-1, 2, and 3) and there
hedgehog proteins may have both direct effects is a degree of cross-reactivity between each
on neurons and also indirect neuroprotective VEGF family member and the VEGFR sub-
effects via regulation of microvascular struc- types such that, for example, VEGF-A binds
ture and function (see above). Vascular endo- both VEGFR-1 and VEGFR-2 while VEGF-D
140 N. A. Calcutt

binds VEGFR-2 and VEGFR-3. The vast major- 8 Erythropoietin


ity of VEGF-­mediated signaling events studied
to date represent VEGF-A binding to VEGFR-2 Erythropoietin (Epo) is a glycoprotein cytokine
with subsequent activation of tyrosine kinases. secreted by renal fibroblasts in response to
In the adult, VEGF-A expression by many cell hypoxia in order to stimulate bone marrow eryth-
types is induced by hypoxia via the transcription ropoiesis. Recombinant Epo is widely used to
factor hypoxia-inducible factor (HIF) and treat anemia in chronic kidney disease and during
secreted VEGF-A binds VEGFR-2 located on cancer therapy. Epo is also induced in multiple
endothelial cells to promote angiogenesis, cell types during periods of stress and has a vari-
increase vascular permeability, and regulate ety of other actions including promoting cell sur-
multiple physiological functions throughout the vival and angiogenesis. Efficacy of Epo extends
body [287]. to direct effects on neurons, as demonstrated by
A role for VEGF in diabetic neuropathy, be it in  vitro studies of cholinergic CNS neurons
pathogenic or reparative, is suggested by reports [302], neuroblastomas [303], and sensory neu-
that certain gene polymorphisms associate with rons of the DRG [304]. Epo triggers erythropoi-
susceptibility to neuropathy in diabetic patients esis via the high affinity Epo receptor (EpoR)
[288–290]. In preclinical studies, diabetes homodimer but can also bind a lower affinity het-
results in transient expression of mRNA for HIF erodimer comprising EpoR and β common recep-
and its target genes, including VEGF [291], tor (βcR) sub-units (the innate repair receptor:
whereas expression of PDGF is reduced [67]. IRR) that is proposed to activate tissue protection
Increased expression of VEGF-A occurs in both and repair [305]. EPO signals in the CNS via
axons and Schwann cells of the PNS and was JAK2/STAT and PI3K/Akt, ERK/MAPK and
prevented by treatment with insulin or NGF other pathways that are associated with neuro-
[292, 293]. This diabetes-induced increase in protection [306]. In the PNS, Epo is present in
VEGF expression could represent an angiogenic around 30% of DRG neuronal cell bodies of all
response to injury/hypoxia but may also con- diameters, axons, and Schwann cell cytoplasm
tribute to neuropathy by promoting vascular and is upregulated in Schwann cells after physi-
leakage and there is evidence to support both cal nerve injury, while EpoR is expressed by neu-
positive and negative effects of VEGF on periph- ronal cell bodies in the DRG, Schwann cells, and
eral nerve. The VEGF-­ sequestering antibody endothelial cells [307]. Proposed functions of
bevacizumab, which is FDA approved as an this EPO/EpoR network in the PNS include neu-
anti-angiogenic agent to treat assorted cancers, ronal survival and regulation of Schwann cell
prevented and reversed indices of neuropathy proliferation following nerve injury [308, 309]
and neuropathic pain in diabetic rats while and development of the neuromuscular junction
in  vitro studies suggested that Schwann cell [303].
derived VEGF contributed to axonal neuropathy Initial studies to determine therapeutic effi-
[294]. Conversely, increasing local VEGF-A cacy in diabetic rodents used recombinant EPO
production by either gene transfer [295–297] or [310–312] or HSV-mediated gene transfer [313,
gene transfer of a VEGF transcription enhancer 314] both of which produced broad efficacy
[298, 299] ameliorated multiple measures of against indices of peripheral neuropathy. As the
neuropathy and neuropathic pain in diabetic neurotrophic properties of Epo are localized to a
rats. Splice variants of VEGF-A have been particular region of the peptide sequence, modi-
reported to either cause (VEGF-A165a) or allevi- fied EPO can be produced that retains neuro-
ate (VEGF-A165b) indices of neuropathic pain in trophic properties but lacks erythropoietic
models of traumatic nerve injury [300], while capacity [303, 315, 316]. This allows targeted
recombinant VEGF-A165b ameliorated indices of neurotrophic activity while minimizing concerns
neuropathy and neuropathic pain in diabetic regarding excess erythropoiesis. EPO derivatives
rodents [301]. lacking erythropoietic activity such as carba-
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 141

mylated EPO and ARA290 (Cibinetide), a modi- fiber neuropathy due to the distribution of their
fied 11-mer sequence of the neurotrophic region receptors, such as NGF [12], or show clinical
of EPO showed efficacy against thermal hypoal- efficacy primarily against small fiber neuropathy,
gesia [316] impaired wound healing [317] and like Epo [320], face the additional obstacle that
autonomic neuropathy [318, 319] in diabetic prevention or reversal of large fiber conduction
rodents. A clinical trial of AR290 given to dia- velocity slowing is currently the de facto primary
betic subjects with neuropathy for 4 weeks, with indicator of efficacy against diabetic neuropathy
a further 4 weeks washout, demonstrated allevia- used by the US FDA. A number of development
tion of pain symptoms [320]. This analgesic programs have been discontinued following lack
property may derive from inhibition of the of efficacy against large fiber conduction slow-
TRPV1 receptor [321] or suppression of spinal ing, despite some, such as BDNF [73] and
microglial activation [322]. Diabetic subjects C-peptide [237] having significant effects on
with the most severe loss of sensory nerves in the other indices of diabetic neuropathy.
cornea also showed recovery of innervation
[320], but more extensive clinical data have yet to Gene transfer  Use of viral vectors to induce
be reported. expression of neurotrophic factors in specific
cells has been employed as a means of removing
the need for repeated systemic injections of
9 Will Neurotrophic Factors recombinant polypeptides [323]. Preclinical
Ever Be a Viable Therapy studies have reported efficacy against multiple
for Diabetic Neuropathy? indices of neuropathy in diabetic rodents after
inducing expression of NGF [39], NT-3 [324,
The discovery that NGF was not the sole factor 325], IGF-1 [326, 327], erythropoietin [313, 314]
that provided neurotrophic support to the PNS, or VEGF [295]. A clinical trial using gene ther-
and technological advances that allowed synthe- apy to increase local VEGF in diabetic patients
sis of large quantities of recombinant peptides with neuropathy reported efficacy against symp-
with appropriate human sequences, generated tom scores, pain, and sensory loss that impact
much initial enthusiasm for using neurotrophic quality of life after 6 months [328]. However,
factors as biologically rational and targeted treat- electrophysiological measures of large fiber neu-
ments for diabetic neuropathy, and indeed many ropathy did not improve and a number of serious
other neurodegenerative diseases. A summary of adverse events were reported [328], perhaps
neurotrophic factors used in clinical trials against reflecting problems inherent to using a factor that
diabetic neuropathy is shown in Table 1. However, has diverse effects in multiple tissues. A separate
the growing appreciation of the diversity of neu- trial involving gene transfer of a VEGF transcrip-
rotrophic and growth factors that act on the PNS tion enhancer also failed to convince in subjects
was accompanied by recognition that individual with diabetic neuropathy [329]. VEGF treatment
factors were less selective for specific popula- in subjects with diabetes is controversial as anti-
tions of neurons than anticipated. Consequently, bodies that bind and neutralize VEGF are used to
initial clinical trials of factors that relied on sys- treat diabetic retinopathy and macular edema
temic injections of recombinant peptide faced not [330, 331]. Moreover, reliance on a single factor
only challenges in delivery and dosing [36], but to correct diverse aspects of neuropathy may be
also concerns regarding potential side effect pro- optimistic. A novel variant of the gene therapy
files. The clinical trial of recombinant CNTF in approach was recently reported in which skin
subjects with motor neuron disease [175] and fibroblasts of adult diabetic mice were repro-
preclinical studies with a hedgehog agonist grammed into cells expressing neuron-like mark-
(Fig. 3) illustrate the problems arising from unan- ers via nano transfection of genes that code for a
ticipated systemic effects of neurotrophic factor range of transcription factors, with the intent of
treatment. Neurotrophic factors that target small modifying the neurotrophic environment of the
142 N. A. Calcutt

Table 1  Clinical trials of neurotrophic and other factors against diabetic neuropathy
Factor Formulation Efficacy vs. Comments Status References
NGF Recombinant Injection site Discontinued Apfel [36]
protein hyperalgesia
Anti-NGF Pain scores Recent data In development Bramson [53]
(tanezumab) (NCT01087203)
BDNF Recombinant Cold detection Injection site Discontinued Wellmer [73]
protein lesions,
increased gut
motility
C-peptide Modified peptide Vibration Placebo effect Discontinued Wahren [237]
perception on NCV
threshold obscured
efficacy
VEGF Gene therapy Pain, sensory Multiple major Discontinued Ropper [328]
loss AE
Transcription Not reported Not reported Discontinued Eisenstein [329]
enhancer
Anti-VEGF Diabetic FDA Approved Glassman [330],
(ranibizumab) retinopathy/ (2015) Striglia [331]
macula edema
Epo Neurotrophic Pain, corneal Small In development Brines [320]
peptide fragment nerve loss exploratory
(Cibinetide) study

epidermis [332]. Nano transfection increased properties when secreted in its uncleaved form
skin NGF and NT-3 mRNA plus NGF protein [333]. The neurotrophic peptide sequence derived
level and was accompanied by an increase in der- from prosaposin was effective against multiple
mal/epidermal PGP9.5 +ve staining which iden- indices of neuropathy in diabetic rodents [334–
tifies neurons and Langerhans cells. Although 339] but a failed clinical trial against neuropathic
gene therapy remains an advance on systemic pain in patients with HIV halted clinical develop-
delivery of single recombinant factors, particu- ment before it could be tested against diabetic neu-
larly where multiple factors can be induced and ropathy [340]. Epo also contains a neurotrophic
localized, it also comes with its own challenges, sequence [303] that has been manipulated to
including how to regulate dosing and turn off fac- develop a neuroprotective therapeutic that lacks
tor production as needed, that will need to be erythropoietic activity and is in clinical develop-
addressed for effective translation to clinical use. ment [316, 320]. Neurotrophic sequences located
in multiple other biologically active peptides and
proteins were identified using the same algorithm
Manipulating the native peptide sequence  One that was applied to prosaposin and Epo but have
approach to avoiding unwanted biological effects not yet been pursued. Another intriguing finding is
of therapeutic peptides is emerging from the recog- the discovery of trophic factor mutants or splice
nition that biological proteins and peptides may variants with distinct biological properties. For
contain otherwise unanticipated neurotrophic example, studies of the NGFR100W mutation associ-
sequences that can be isolated from the biological ated with loss of pain sensation in Hereditary
effects associated with the parent molecule. An Sensory and Autonomic Neuropathy type V, have
early example was the discovery of a neurotrophic helped separate the neurotrophic and algogenic
region in prosaposin, which is a precursor of the properties of the NGF molecule [341–344] and
lysosomal saposins that facilitate catabolism of may allow development of a “pain free” NGF ther-
glycosphingolipds and also shows neurotrophic apy. Other non-algogenic NGF mutants have also
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 143

been identified [46] and accelerate wound healing lating intrinsic growth and repair mechanisms
and reinnervation in diabetic mice [345]. VEGF-A that are distant from specific neurotrophic factors
also exists as splice variants that have been reported and their immediate receptor signaling cascades
to cause (VEGF-A165a) or alleviate (VEGF-A165b) and may be common to all neuronal sub-­
indices of neuropathic pain in models of traumatic populations. A strong case has been made that
nerve injury [300]. Recombinant VEGF-A165b ame- nerve growth and repair is tonically restrained by
liorated indices of neuropathy and neuropathic pain tumor suppressors such as PTEN, Rb1, and APC
in diabetic rodents [301], suggesting that this vari- and that lifting this restraint may allow nerve
ant may have therapeutic potential. Perhaps most regeneration and perhaps neuroprotection [8].
intriguingly, the balance between endogenous Evidence of efficacy against diabetic neuropathy
VEGF-­A165a and VEGF-A165b can be manipulated is currently limited to a study in which PTEN
by modulators of alternative splicing [346, 347] knockdown improved regeneration after crush
and could lead to a small molecule enhancer of injury in the nerve of diabetic mice [348]. Sensory
therapeutic VEGF-A165b isoform. neuron regenerative growth is also under tonic
constraint mediated by cholinergic signaling via
muscarinic M1 receptors that depresses mito-
Manipulating neurotrophic signaling  Small chondrial function, and cellular energy produc-
molecule inducers of endogenous neurotrophic tion [349–351]. While it is not clear that excess
factor production may have a better safety profile cholinergic constraint is a contributory mecha-
than exogenously delivered factors and dosing nism to diabetic neuropathy, removal of the
can be better regulated than gene therapy. The intrinsic cholinergic constraint on energy produc-
molecules studied to date tend to have a multi- tion using muscarinic antagonists prevents and
tude of biological effects outside their induction reverses multiple indices of neuropathy in dia-
of neurotrophic support and it is not yet clear betic rodents [352, 353]. Further preclinical and
which are responsible for the beneficial effects on clinical studies of the therapeutic potential of
neuropathy in diabetic rodents—although modulating intrinsic neuronal growth restraint
impacting a variety of potential pathogenic mechanisms are in progress.
mechanisms is not in itself undesirable and may
ultimately prove a more pragmatic approach to
treating a condition that may have multiple con- Stem cells and secretomes  The impact of dia-
tributing lesions. The growing understanding of betes on all components of the PNS presents
the molecular biology of the receptors and sig- problems for the therapeutic development of neu-
naling pathways associated with neurotrophic rotrophic factors that target specific sub-­
factors has also allowed design of small molecule populations of neurons. One approach has been
mimetics, agonists, and modulators of down- to deliver cocktails of factors that together cover
stream signaling pathways that it is hoped will a broad spectrum of neurons, such as the combi-
obviate the need for peptide injections and allow nation of NGF and bFGF to enhance nerve regen-
easily regulated delivery. Examples include eration in diabetic rats [283]. However, this is not
BDNF mimetics [76, 77], both RET agonists always beneficial, for example, the addition of
[138] and modulators of RET signaling [139] to NT-3 to NGF diminished efficacy of NGF possi-
mimic GDNF bioactivity, and a Smo agonist that bly by competing with NGF for the p75NTR [118].
replicates the consequences of hedgehog binding Reprogramming keratinocytes to create a local
to patched [279] although, as illustrated in Fig. 3, environment in the skin that is rich in multiple
they remain susceptible to adverse events and neurotrophic factors has been discussed above
side effects depending on the profile of receptor [332]. Another promising development has been
distribution and biological function. Most use of stem cells, not to rebuild peripheral nerve
recently, there is an interest in assuming the bio- by engraftment and differentiation but rather to
logical roles of neurotrophic factors by manipu- act as paracrine delivery systems for a cocktail of
144 N. A. Calcutt

factors, their secretome, which may not only pro- 3. LeviMontalcini R, et  al. Nerve growth factor:
vide neurotrophic support but could also impact from neurotrophin to neurokine. Trends Neurosci.
1996;19(11):514–20.
other potential pathogenic mechanisms associ- 4. Scott-Solomon E, Kuruvilla R. Mechanisms of neu-
ated with diabetic neuropathy, such as rebalanc- rotrophin trafficking via Trk receptors. Mol Cell
ing of a pro-inflammatory environment and repair Neurosci. 2018;91:25–33.
of the microvasculature and/or blood:nerve bar- 5. Thomas PK.  Classification, differential diagno-
sis, and staging of diabetic peripheral neuropathy.
rier [354]. Studies using stem cells from diverse Diabetes. 1997;46:S54–7.
sources that were injected into diabetic rodents 6. Goncalves NP, et al. Schwann cell interactions with
have shown that efficacy to alleviate neuropathy axons and microvessels in diabetic neuropathy. Nat
was accompanied by secretion of an array of fac- Rev Neurol. 2017;13(3):135–47.
7. Dong J, et  al. Mast cells in diabetes and diabetic
tors including NGF, NT3, bFGF, and VEGF that wound healing. Adv Ther. 2020;37(11):4519–37.
together can potentially impact a broad cross sec- 8. Duraikannu A, et al. Beyond trophic factors: exploit-
tion of cells associated with the PNS, while the ing the intrinsic regenerative properties of adult neu-
stem cells themselves may also differentiate into rons. Front Cell Neurosci. 2019;13:128.
9. Nowak NC, et  al. Cutaneous innervation in
location appropriate cell types [355–357]. In one impaired diabetic wound healing. Transl Res.
study, the improvement in nerve blood flow and 2021;236:87–108.
conduction velocity seen in diabetic rats treated 10. Hempstead BL.  Deciphering proneurotrophin
with bone marrow or blood derived mononuclear actions. Handb Exp Pharmacol. 2014;220:17–32.
11. Marchetti L, et  al. Fast-diffusing p75(NTR) mono-
cells was blocked by anti-VEGF antibodies mers support apoptosis and growth cone collapse
[358]. Adipose-derived stem cells (ASC’s) appear by neurotrophin ligands. Proc Natl Acad Sci U S A.
to show particular promise as their secretome 2019;116(43):21563–72.
contains diverse neurotrophic and immunomodu- 12. Fang X, et al. trkA is expressed in nociceptive neu-
rons and influences electrophysiological properties
latory factors [359] that act on adjacent cells in a via Nav1.8 expression in rapidly conducting noci-
paracrine manner. For example, ASC’s delivered ceptors. J Neurosci. 2005;25(19):4868–78.
to the ends of transected nerves secrete multiple 13. Lindsay RM.  Nerve growth-factors (Ngf, Bdnf)
neurotrophic factors, differentiate into Schwann enhance axonal regeneration but are not required
for survival of adult sensory neurons. J Neurosci.
cells and enhance peripheral nerve repair [179]. 1988;8(7):2394–405.
In preclinical studies, both human ASC’s and 14. Lindsay RM, Harmar AJ. Nerve growth-factor regu-
fluid containing their secretions reversed indices lates expression of neuropeptide genes in adult sen-
of neuropathic pain and prevented loss of IENF sory neurons. Nature. 1989;337(6205):362–4.
15. Bennett DLH, et al. Endogenous nerve growth factor
in diabetic mice [190] while ASC’s may also regulates the sensitivity of nociceptors in the adult
impact both the vascular and neurological con- rat. Eur J Neurosci. 1998;10(4):1282–91.
tributors to erectile dysfunction in diabetic 16. Chudler EH, Anderson LC, Byers MR.  Nerve
rodents [360]. Given the relative ease of harvest- growth factor depletion by autoimmunization pro-
duces thermal hypoalgesia in adult rats. Brain Res.
ing ASC’s and their secretome, the potential for 1997;765(2):327–30.
returning ASC’s as an allograft to treat diabetic 17. Barker PA, et al. Nerve growth factor signaling and
patients is an intriguing extension of the use of its contribution to pain. J Pain Res. 2020;13:1223–41.
neurotrophic factors as therapies for diabetic 18. Lewin GR, Ritter AM, Mendell LM. Nerve growth-­
factor induced hyperalgesia in the neonatal and
neuropathy that deserves further exploration. adult-rat. J Neurosci. 1993;13(5):2136–48.
19. Petty BG, et  al. The effect of systemically
administered recombinant human nerve growth-­
factor in healthy-human subjects. Ann Neurol.
1994;36(2):244–6.
References 20. Fernyhough P, et  al. Deficits in sciatic nerve neu-
ropeptide content coincide with a reduction in tar-
1. Howe CL, Mobley WC.  Long-distance retrograde get tissue nerve growth factor messenger RNA in
neurotrophic signaling. Curr Opin Neurobiol. streptozotocin-­diabetic rats: effects of insulin treat-
2005;15(1):40–8. ment. Neuroscience. 1994;62(2):337–44.
2. Levi-Montalcini R. The nerve growth factor 35 years 21. Fernyhough P, et  al. Altered neurotrophin mRNA
later. Science. 1987;237(4819):1154–62. levels in peripheral nerve and skeletal muscle
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 145

of experimentally diabetic rats. J Neurochem. went right, and what does the future hold? Int Rev
1995;64(3):1231–7. Neurobiol. 2002;50:393–413.
22. Fernyhough P, et  al. Human recombinant nerve 37. Zhao M, et al. Efficacy and safety of nerve growth
growth factor replaces deficient neurotrophic factor for the treatment of neurological diseases:
support in the diabetic rat. Eur J Neurosci. a meta-analysis of 64 randomized controlled tri-
1995;7(5):1107–10. als involving 6,297 patients. Neural Regen Res.
23. Hellweg R, Hartung HD.  Endogenous levels of 2015;10(5):819–28.
nerve growth-factor (Ngf) are altered in experimen- 38. Wu YX, et  al. Nerve growth factor improves the
tal diabetes-mellitus—a possible role for Ngf in the outcome of type 2 diabetes-induced hypotestos-
pathogenesis of diabetic neuropathy. J Neurosci Res. teronemia and erectile dysfunction. Reprod Sci.
1990;26(2):258–67. 2019;26(3):386–93.
24. Brewster WJ, et  al. Reduced sciatic nerve sub- 39. Walwyn WM, et al. HSV-1-mediated NGF delivery
stance P and calcitonin gene-related peptide in delays nociceptive deficits in a genetic model of dia-
rats with short-term diabetes or central hypoxae- betic neuropathy. Exp Neurol. 2006;198(1):260–70.
mia co-exist with normal messenger RNA levels 40. Tsafack EG, et al. Antihypernociceptive and neuro-
in the lumbar dorsal root ganglia. Neuroscience. protective effects of the aqueous and methanol stem-­
1994;58(2):323–30. bark extracts of Nauclea pobeguinii (Rubiaceae) on
25. Calcutt NA, et  al. Axonal transport of substance STZ-induced diabetic neuropathic pain. Evid Based
P-like immunoreactivity in ganglioside-treated dia- Complement Alternat Med. 2021;2021:6637584.
betic rats. J Neurol Sci. 1990;96(2–3):283–91. 41. Riaz S, et  al. A vitamin D-3 derivative (CB1093)
26. Diemel LT, et  al. Expression of neuropeptides in induces nerve growth factor and prevents neu-
experimental diabetes; effects of treatment with nerve rotrophic deficits in streptozotocin-diabetic rats.
growth factor or brain-derived neurotrophic factor. Diabetologia. 1999;42(11):1308–13.
Brain Res Mol Brain Res. 1994;21(1–2):171–5. 42. Calcutt NA, et  al. Protection of sensory function
27. Tomlinson DR, Fernyhough P, Diemel LT.  Role of in diabetic rats by Neotrofin. Eur J Pharmacol.
neurotrophins in diabetic neuropathy and treatment 2006;534(1–3):187–93.
with nerve growth factors. Diabetes. 1997;46(Suppl 43. Hanaoka Y, et  al. The therapeutic effects of
2):S43–9. 4-­methylcatechol, a stimulator of endogenous nerve
28. Robinson JP, et  al. Axonal transport and tis- growth-factor synthesis, on experimental diabetic
sue contents of substance P in rats with long- neuropathy in rats. J Neurol Sci. 1994;122(1):28–32.
term streptozotocin-­ diabetes. Effects of the 44. Comelli F, et  al. Rimonabant, a cannabinoid CB1
aldose reductase inhibitor ‘statil’. Brain Res. receptor antagonist, attenuates mechanical allodynia
1987;426(2):339–48. and counteracts oxidative stress and nerve growth
29. Bennett GS, et al. Neurogenic cutaneous vasodilata- factor deficit in diabetic mice. Eur J Pharmacol.
tion and plasma extravasation in diabetic rats: effect 2010;637(1–3):62–9.
of insulin and nerve growth factor. Br J Pharmacol. 45. Wang L, et  al. Tadalafil promotes the recovery of
1998;124(7):1573–9. peripheral neuropathy in type II diabetic mice. PLoS
30. Calcutt NA. Experimental models of painful diabetic One. 2016;11(7):e0159665.
neuropathy. J Neurol Sci. 2004;220(1–2):137–9. 46. Malerba F, et  al. Functional characterization of
31. Calcutt NA, et al. Elevated substance-P-like immu- human ProNGF and NGF mutants: identification
noreactivity levels in spinal dialysates during the of NGF P61SR100E as a “painless” lead investiga-
formalin test in normal and diabetic rats. Brain Res. tional candidate for therapeutic applications. PLoS
2000;856(1–2):20–7. One. 2015;10(9):e0136425.
32. Garrett NE, et al. alpha-Lipoic acid corrects neuro- 47. Ro LS, et  al. Effect of NGF and anti-NGF
peptide deficits in diabetic rats via induction of tro- on neuropathic pain in rats following chronic
phic support. Neurosci Lett. 1997;222(3):191–4. constriction injury of the sciatic nerve. Pain.
33. Calcutt NA, Chen P, Hua XY.  Effects of diabetes 1999;79(2–3):265–74.
on tissue content and evoked release of calcitonin 48. Sevcik MA, et  al. Anti-NGF therapy profoundly
gene-related peptide-like immunoreactivity from rat reduces bone cancer pain and the accompanying
sensory nerves. Neurosci Lett. 1998;254(3):129–32. increase in markers of peripheral and central sensiti-
34. Sango K, et al. Nerve growth factor (NGF) restores zation. Pain. 2005;115(1–2):128–41.
depletions of calcitonin gene-related peptide and 49. Dietz BW, et  al. Targeting nerve growth fac-
substance P in sensory neurons from diabetic mice tor for pain management in osteoarthritis-clinical
in vitro. J Neurol Sci. 1994;126(1):1–5. efficacy and safety. Rheum Dis Clin North Am.
35. Unger JW, et al. Nerve growth factor (NGF) and dia- 2021;47:181–95.
betic neuropathy in the rat: morphological investi- 50. Doshi TL, Tesarz J, Cohen SP. Nerve growth factor
gations of the sural nerve, dorsal root ganglion, and inhibitors for low back pain: balancing the risks and
spinal cord. Exp Neurol. 1998;153(1):23–34. benefits. Pain. 2020;161(9):1941–2.
36. Apfel SC.  Nerve growth factor for the treatment 51. Markman JD, et  al. Tanezumab for chronic low
of diabetic neuropathy: what went wrong, what back pain: a randomized, double-blind, placebo-
146 N. A. Calcutt

and active-controlled, phase 3 study of efficacy and neurotrophic factor in diabetic rats. Curr Neurovasc
safety. Pain. 2020;161(9):2068–78. Res. 2010;7(3):213–22.
52. Patel MK, Kaye AD, Urman RD. Tanezumab: therapy 67. Wang Q, et al. Danhong injection alleviates mechan-
targeting nerve growth factor in pain pathogenesis. J ical allodynia via inhibiting ERK1/2 activation
Anaesthesiol Clin Pharmacol. 2018;34(1):111–6. and elevates BDNF level in sciatic nerve in dia-
53. Bramson C, et al. Exploring the role of tanezumab as betic rat. Evid Based Complement Alternat Med.
a novel treatment for the relief of neuropathic pain. 2018;2018:5798453.
Pain Med. 2015;16(6):1163–76. 68. Zhou GJ, et  al. Ameliorative effect of berberine
54. Nitta A, et  al. Diabetic neuropathies in brain are on neonatally induced type 2 diabetic neuropathy
induced by deficiency of BDNF.  Neurotoxicol via modulation of BDNF, IGF-1, PPAR-gamma,
Teratol. 2002;24(5):695–701. and AMPK expressions. Dose Response.
55. Kim OY, Song J.  The importance of BDNF and 2019;17(3):1559325819862449.
RAGE in diabetes-induced dementia. Pharmacol 69. Zhang CH, et  al. The Akt/mTOR cascade medi-
Res. 2020;160:105083. ates high glucose-induced reductions in BDNF via
56. Afarid M, Namvar E, Sanie-Jahromi F.  Diabetic DNMT1 in Schwann cells in diabetic peripheral neu-
retinopathy and BDNF: a review on its molecu- ropathy. Exp Cell Res. 2019;383(1):111502.
lar basis and clinical applications. J Ophthalmol. 70. Rodriguez-Pena A, et  al. Expression of neu-
2020;2020:1602739. rotrophins and their receptors in sciatic nerve
57. Richner M, et al. Peripheral nerve injury modulates of experimentally diabetic rats. Neurosci Lett.
neurotrophin signaling in the peripheral and central 1995;200(1):37–40.
nervous system. Mol Neurobiol. 2014;50(3):945–70. 71. Mizisin AP, et al. Decreased accumulation of endog-
58. Garcia N, et  al. Localization of brain-derived neu- enous brain-derived neurotrophic factor against
rotrophic factor, neurotrophin-4, tropomyosin- constricting sciatic nerve ligatures in streptozotocin-­
related kinase b receptor, and p75NTR receptor by diabetic and galactose-fed rats. Neurosci Lett.
high-­resolution immunohistochemistry on the adult 1999;263(2–3):149–52.
mouse neuromuscular junction. J Peripher Nerv 72. Calcutt NA, Freshwater JD, Mizisin AP. Prevention
Syst. 2010;15(1):40–9. of sensory disorders in diabetic Sprague-Dawley
59. Garraway SM, Petruska JC, Mendell LM.  BDNF rats by aldose reductase inhibition or treatment
sensitizes the response of lamina II neurons to high with ciliary neurotrophic factor. Diabetologia.
threshold primary afferent inputs. Eur J Neurosci. 2004;47(4):718–24.
2003;18(9):2467–76. 73. Wellmer A, et  al. A double-blind placebo-
60. Kerr BJ, et  al. Brain-derived neurotrophic factor controlled clinical trial of recombinant human
modulates nociceptive sensory inputs and NMDA-­ brain-derived neurotrophic factor (rhBDNF) in
evoked responses in the rat spinal cord. J Neurosci. diabetic polyneuropathy. J Peripher Nerv Syst.
1999;19(12):5138–48. 2001;6(4):204–10.
61. Lever IJ, et  al. Brain-derived neurotrophic fac- 74. Ge HX, et al. Dihydromyricetin affects BDNF lev-
tor is released in the dorsal horn by distinctive els in the nervous system in rats with comorbid
patterns of afferent fiber stimulation. J Neurosci. diabetic neuropathic pain and depression. Sci Rep.
2001;21(12):4469–77. 2019;9:14619.
62. Slack SE, et  al. Brain-derived neurotrophic factor 75. Maekawa T, et al. Synthesis and biological activity
induces NMDA receptor subunit one phosphoryla- of novel 5-(omega-aryloxyalkyl)oxazole derivatives
tion via ERK and PKC in the rat spinal cord. Eur J as brain-derived neurotrophic factor inducers. Chem
Neurosci. 2004;20(7):1769–78. Pharm Bull. 2003;51(5):565–73.
63. Copray S, et  al. Contraction-induced muscle 76. Fobian K, et  al. Peptides derived from the
fiber damage is increased in soleus muscle of solvent-­
exposed loops 3 and 4 of BDNF bind
streptozotocin-­diabetic rats and is associated with TrkB and p75(NTR) receptors and stimulate
elevated expression of brain-derived neurotrophic neurite outgrowth and survival. J Neurosci Res.
factor mRNA in muscle fibers and activated satellite 2010;88(6):1170–81.
cells. Exp Neurol. 2000;161(2):597–608. 77. Yang SH, et  al. Self-assembling peptide hydrogels
64. Mizisin AP, et  al. BDNF attenuates functional and functionalized with LN- and BDNF- mimicking
structural disorders in nerves of galactose-fed rats. epitopes synergistically enhance peripheral nerve
J Neuropathol Exp Neurol. 1997;56(12):1290–301. regeneration. Theranostics. 2020;10(18):8227–49.
65. Fernyhough P, Maeda K, Tomlinson DR.  Brain-­ 78. Lee-Kubli CAG, Calcutt NA. Altered rate-­dependent
derived neurotrophic factor mRNA levels are depression of the spinal H-reflex as an indicator of
up-regulated in hindlimb skeletal muscle of dia- spinal disinhibition in models of neuropathic pain.
betic rats: effect of denervation. Exp Neurol. Pain. 2014;155(2):250–60.
1996;141(2):297–303. 79. Morgado C, et  al. Minocycline completely
66. Li RL, et al. Angelica injection promotes peripheral reverses mechanical hyperalgesia in diabetic rats
nerve structure and function recovery with increased through microglia-induced changes in the expres-
expressions of nerve growth factor and brain derived sion of the potassium chloride co-transporter 2
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 147

(KCC2) at the spinal cord. Diabetes Obes Metab. rate-dependent depression of the Hoffmann reflex in
2011;13(2):150–9. diabetic neuropathic pain and reduce primary affer-
80. Ferrini F, De Koninck Y. Microglia control neuronal ent excitability. Pain. 2019;160(6):1448–58.
network excitability via BDNF signalling. Neural 96. Lee-Kubli CA, et  al. Pharmacological modulation
Plast. 2013;2013:429815. of rate-dependent depression of the spinal H-reflex
81. Tsuda M.  Microglia-mediated regulation of neu- predicts therapeutic efficacy against painful diabetic
ropathic pain: molecular and cellular mechanisms. neuropathy. Diagnostics. 2021;11(2):283.
Biol Pharm Bull. 2019;42(12):1959–68. 97. Marshall AG, et  al. Spinal disinhibition in experi-
82. Watkins LR, Maier SF. When good pain turns bad. mental and clinical painful diabetic neuropathy.
Curr Dir Psychol Sci. 2003;12(6):232–6. Diabetes. 2017;66(5):1380–90.
83. Malcangio M, Montague K. Neuroimmune commu- 98. Worthington A, et al. Spinal inhibitory dysfunction
nication in chemotherapy-induced neuropathic pain. in patients with painful or painless diabetic neuropa-
Glia. 2017;65:E96–7. thy. Diabetes Care. 2021;44(8):1835–41.
84. Cho HJ, et al. Changes in brain-derived neurotrophic 99. Barbacid M. The Trk family of neurotrophin recep-
factor immunoreactivity in rat dorsal root ganglia, tors. J Neurobiol. 1994;25(11):1386–403.
spinal cord, and gracile nuclei following cut or crush 100. Lindsay RM.  Role of neurotrophins and trk recep-
injuries. Exp Neurol. 1998;154(1):224–30. tors in the development and maintenance of sensory
85. Mannion RJ, et  al. Neurotrophins: peripherally neurons: an overview. Philos Trans R Soc Lond B
and centrally acting modulators of tactile stimulus-­ Biol Sci. 1996;351(1338):365–73.
induced inflammatory pain hypersensitivity. Proc 101. Lewis MA, et  al. Identification and character-
Natl Acad Sci U S A. 1999;96(16):9385–90. ization of compounds that potentiate NT-3-­
86. Lee-Kubli C, et al. The H-reflex as a biomarker for mediated Trk receptor activity. Mol Pharmacol.
spinal disinhibition in painful diabetic neuropathy. 2006;69(4):1396–404.
Curr Diab Rep. 2018;18(1):1. 102. Distefano PS, et  al. The neurotrophins Bdnf, Nt-3,
87. Miao B, et al. The implication of transient receptor and Ngf display distinct patterns of retrograde
potential canonical 6 in BDNF-induced mechanical axonal-transport in peripheral and central neurons.
allodynia in rat model of diabetic neuropathic pain. Neuron. 1992;8(5):983–93.
Life Sci. 2021;273:119308. 103. Altar CA, DiStefano PS.  Neurotrophin traffick-
88. Daulhac L, et al. Phosphorylation of spinal N-methyl-­ ing by anterograde transport. Trends Neurosci.
D-aspartate receptor NR1 subunits by extracellular 1998;21(10):433–7.
signal-regulated kinase in dorsal horn neurons and 104. von Bartheld CS, Wang XX, Butowt R. Anterograde
microglia contributes to diabetes-induced painful axonal transport, transcytosis, and recycling of
neuropathy. Eur J Pain. 2011;15(2):169.e1–169.e12. neurotrophic factors—the concept of trophic
89. Rivera C, et  al. BDNF-induced TrkB activation currencies in neural networks. Mol Neurobiol.
down-regulates the K+-Cl- cotransporter KCC2 2001;24(1–3):1–28.
and impairs neuronal Cl- extrusion. J Cell Biol. 105. Naito Y, Lee AK, Takahashi H. Emerging roles of the
2002;159(5):747–52. neurotrophin receptor TrkC in synapse organization.
90. Rivera C, Voipio J, Kaila K.  Two developmental Neurosci Res. 2017;116:10–7.
switches in GABAergic signalling: the K+-Cl- 106. Boyce VS, Mendell LM.  Neurotrophins and spinal
cotransporter KCC2 and carbonic anhydrase circuit function. Front Neural Circuits. 2014;8:59.
CAVII. J Physiol. 2005;562(1):27–36. 107. Andreassen CS, et  al. Expression of neurotrophic
91. Miletic G, Miletic V.  Loose ligation of the sciatic factors in diabetic muscle-relation to neuropathy and
nerve is associated with TrkB receptor-dependent muscle strength. Brain. 2009;132:2724–33.
decreases in KCC2 protein levels in the ipsilateral 108. Fernyhough P, Diemel LT, Tomlinson DR.  Target
spinal dorsal horn. Pain. 2008;137(3):532–9. tissue production and axonal transport of neuro-
92. Jolivalt CG, et  al. Allodynia and hyperalgesia in trophin-­3 are reduced in streptozotocin-diabetic rats.
diabetic rats are mediated by GABA and depletion Diabetologia. 1998;41(3):300–6.
of spinal potassium-chloride co-transporters. Pain. 109. Ihara C, et al. Decreased neurotrophin-3 expression
2008;140(1):48–57. in skeletal muscles of streptozotocin-induced dia-
93. Morgado C, Pinto-Ribeiro F, Tavares I.  Diabetes betic rats. Neuropeptides. 1996;30(4):309–12.
affects the expression of GABA and potassium 110. Cai F, Tomlinson DR, Fernyhough P.  Elevated
chloride cotransporter in the spinal cord: a study expression of neurotrophin-3 mRNA in sensory
in streptozotocin diabetic rats. Neurosci Lett. nerve of streptozotocin-diabetic rats. Neurosci Lett.
2008;438(1):102–6. 1999;263(2–3):81–4.
94. Boulenguez P, et  al. Down-regulation of the 111. Kennedy AJ, et  al. Neurotrophin-3 is increased
potassium-­ chloride cotransporter KCC2 contrib- in skin in human diabetic neuropathy. J Neurol
utes to spasticity after spinal cord injury. Nat Med. Neurosurg Psychiatry. 1998;65(3):393–5.
2010;16(3):302–7. 112. Terenghi G, et  al. trkA and trkC expression is
95. Hernandez-Reyes JE, et  al. alpha(5)GABA(A) increased in human diabetic skin. Neurosci Lett.
receptors play a pronociceptive role and avoid the 1997;228(1):33–6.
148 N. A. Calcutt

113. Mizisin AP, et al. Neurotrophin-3 reverses nerve con- opmental dependence on ret signaling. Neuron.
duction velocity deficits in streptozotocin-diabetic 2009;64(6):841–56.
rats. J Peripher Nerv Syst. 1999;4(3–4):211–21. 129. Luo WQ, et  al. A hierarchical NGF signaling cas-
114. Mizisin AP, et  al. NT-3 attenuates functional and cade controls Ret-dependent and Ret-independent
structural disorders in sensory nerves of galactose-fed events during development of nonpeptidergic DRG
rats. J Neuropathol Exp Neurol. 1998;57(9):803–13. neurons. Neuron. 2007;54(5):739–54.
115. Middlemas A, et  al. Enhanced activation of axo- 130. Molliver DC, et al. IB4-binding DRG neurons switch
nally transported stress-activated protein kinases in from NGF to GDNF dependence in early postnatal
peripheral nerve in diabetic neuropathy is prevented life. Neuron. 1997;19(4):849–61.
by neurotrophin-3. Brain. 2003;126:1671–82. 131. Bar KJ, et al. GDNF and its receptor component Ret
116. Schmidt RE, et  al. Effect of NGF and neuro- in injured human nerves and dorsal root ganglia.
trophin-­3 treatment on experimental diabetic auto- Neuroreport. 1998;9(1):43–7.
nomic neuropathy. J Neuropathol Exp Neurol. 132. Hoke A, et  al. A decline in glial cell-line-derived
2001;60(3):263–73. neurotrophic factor expression is associated with
117. Li HW, et al. Muscle NT-3 levels increased by exer- impaired regeneration after long-term Schwann cell
cise training contribute to the improvement in caudal denervation. Exp Neurol. 2002;173(1):77–85.
nerve conduction velocity in diabetic rats. Mol Med 133. Christianson JA, Riekhof JT, Wright DE. Restorative
Rep. 2012;6(1):69–74. effects of neurotrophin treatment on diabetes-­
118. Christianson JA, et  al. Neurotrophic modulation induced cutaneous axon loss in mice. Exp Neurol.
of myelinated cutaneous innervation and mechani- 2003;179(2):188–99.
cal sensory loss in diabetic mice. Neuroscience. 134. Saudek F, et  al. Preserved expression of skin neu-
2007;145(1):303–13. rotrophic factors in advanced diabetic neuropathy
119. Huang TJ, et  al. Diabetes-induced alterations does not lead to neural regeneration despite pan-
in calcium homeostasis in sensory neurones of creas and kidney transplantation. J Diabetes Res.
streptozotocin-­ diabetic rats are restricted to lum- 2018;2018:2309108.
bar ganglia and are prevented by neurotrophin-3. 135. Liu GS, et  al. Peripheral gene transfer of glial
Diabetologia. 2002;45(4):560–70. cell-derived neurotrophic factor ameliorates neu-
120. Huang TJ, et  al. Neurotrophin-3 prevents mito- ropathic deficits in diabetic rats. Hum Gene Ther.
chondrial dysfunction in sensory neurons 2009;20(7):715–27.
of streptozotocin-­ diabetic rats. Exp Neurol. 136. Akkina SK, Patterson CL, Wright DE.  GDNF res-
2005;194(1):279–83. cues nonpeptidergic unmyelinated primary afferents
121. Sayers NM, et al. Neurotrophin-3 prevents the proxi- in streptozotocin-treated diabetic mice. Exp Neurol.
mal accumulation of neurofilament proteins in sen- 2001;167(1):173–82.
sory neurons of streptozocin-induced diabetic rats. 137. Christianson JA, et  al. Beneficial actions of neuro-
Diabetes. 2003;52(9):2372–80. trophin treatment on diabetes-induced hypoalgesia
122. Sariola H, Saarma M.  Novel functions and in mice. J Pain. 2003;4(9):493–504.
signalling pathways for GDNF.  J Cell Sci. 138. Viisanen H, et al. Novel RET agonist for the treat-
2003;116(19):3855–62. ment of experimental neuropathies. Mol Pain.
123. Paratcha G, Ledda F, Ibanez CF.  The neural cell 2020;16:1744806920950866.
adhesion molecule NCAM is an alternative sig- 139. Hedstrom KL, et  al. Treating small fiber neuropa-
naling receptor for GDNF family ligands. Cell. thy by topical application of a small molecule
2003;113(7):867–79. modulator of ligand-induced GFR alpha/RET
124. Donnelly CR, Pierchala BA.  Plasma membrane receptor signaling. Proc Natl Acad Sci U S A.
localization of the GFL receptor components: 2014;111(6):2325–30.
a nexus for receptor crosstalk. Cell Tissue Res. 140. Boulton TG, Stahl N, Yancopoulos GD.  Ciliary
2020;382(1):57–64. neurotrophic factor leukemia inhibitory fac-
125. Stanga S, Boido M, Kienlen-Campard P.  How to tor interleukin-6 oncostatin-M family of cyto-
build and to protect the neuromuscular junction: the kines induces tyrosine phosphorylation of a
role of the glial cell line-derived neurotrophic factor. common set of proteins overlapping those induced
Int J Mol Sci. 2021;22(1):136. by other cytokines and growth-factors. J Biol Chem.
126. Zahavi EE, et  al. A compartmentalized micro- 1994;269(15):11648–55.
fluidic neuromuscular co-culture system reveals 141. Pasquin S, Sharma M, Gauchat JF.  Cytokines of
spatial aspects of GDNF functions. J Cell Sci. the LIF/CNTF family and metabolism. Cytokine.
2015;128(6):1241–52. 2016;82:122–4.
127. Enomoto H, et  al. RET signaling is essential for 142. Gloaguen I, et  al. Ciliary neurotrophic factor cor-
migration, axonal growth and axon guidance of rects obesity and diabetes associated with leptin
developing sympathetic neurons. Development. deficiency and resistance. Proc Natl Acad Sci U S A.
2001;128(20):3963–74. 1997;94(12):6456–61.
128. Luo WQ, et  al. Molecular identification of rap- 143. Davis S, et al. The receptor for ciliary neurotrophic
idly adapting mechanoreceptors and their devel- factor. Science. 1991;253(5015):59–63.
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 149

144. Schuster B, et  al. Signaling of human ciliary neu- 162. Canclini L, et  al. Association of microtubules
rotrophic factor (CNTF) revisited—the interleukin-6 and axonal RNA transferred from myelinat-
receptor can serve as an alpha-receptor for CNTF. J ing Schwann cells in rat sciatic nerve. PLoS One.
Biol Chem. 2003;278(11):9528–35. 2020;15(5):e0233651.
145. Dobrea GM, Unnerstall JR, Rao MS. The expression 163. Court FA, et  al. Schwann cell to axon transfer of
of Cntf message and immunoreactivity in the central ribosomes: toward a novel understanding of the
and peripheral nervous-system of the rat. Dev Brain role of glia in the nervous system. J Neurosci.
Res. 1992;66(2):209–19. 2008;28(43):11024–9.
146. Rende M, et al. Immunolocalization of ciliary neu- 164. Court FA, et  al. Axonal autonomous and glial-­
ronotrophic factor in adult-rat sciatic-nerve. Glia. dependent processes participate in axonal degenera-
1992;5(1):25–32. tion. Glia. 2011;59:S73.
147. Lee DA, Zurawel RH, Windebank AJ. Ciliary neuro- 165. Duncan A, et al. Transfer of horseradish peroxidase
trophic factor expression in Schwann-cells is induced from oligodendrocyte to axon in the myelinating
by axonal contact. J Neurochem. 1995;65(2):564–8. neonatal rat optic nerve: artefact or transcellular
148. Sendtner M, et al. Brain-derived neurotrophic factor exchange? Glia. 1996;17(4):349–55.
prevents the death of motoneurons in newborn rats 166. Lasek RJ, Gainer H, Barker J. Transfer of newly syn-
after nerve-section. Nature. 1992;360(6406):757–9. thesized proteins from Schwann-cells to squid giant-­
149. Smith GM, et  al. Temporal and spatial expression axon. Anat Rec. 1974;178(2):398.
of ciliary neurotrophic factor after peripheral-nerve 167. Lopez-Leal R, Diaz P, Court FA.  In vitro analysis
injury. Exp Neurol. 1993;121(2):239–47. of the role of Schwann cells on axonal degeneration
150. Gravel C, et  al. Adenoviral gene transfer of ciliary and regeneration using sensory neurons from dorsal
neurotrophic factor and brain-derived neurotrophic root ganglia. Methods Mol Biol. 2018;1739:255–67.
factor leads to long-term survival of axotomized 168. Shakhbazau A, et  al. Demyelination induces trans-
motor neurons. Nat Med. 1997;3(7):765–70. port of ribosome-containing vesicles from glia to
151. Sendtner M, et  al. Effect of ciliary neurotrophic axons: evidence from animal models and MS patient
factor (Cntf) on motoneuron survival. J Cell Sci. brains. Mol Biol Rep. 2016;43(6):495–507.
1991;15:103–9. 169. Sotelo JR, et  al. Glia to axon RNA transfer. Dev
152. Sendtner M, et al. Endogenous ciliary neurotrophic Neurobiol. 2014;74(3):292–302.
factor is a lesion factor for axotomized motoneurons 170. Mizisin AP, et  al. Ciliary neurotrophic fac-
in adult mice. J Neurosci. 1997;17(18):6999–7006. tor improves nerve conduction and ameliorates
153. Friedman B, et al. Regulation of ciliary neurotrophic regeneration deficits in diabetic rats. Diabetes.
factor expression in myelin-related Schwann-cells 2004;53(7):1807–12.
invivo. Neuron. 1992;9(2):295–305. 171. Gao N, et al. Dendritic cell dysfunction and diabetic
154. Godinho MJ, et al. Regeneration of adult rat sensory sensory neuropathy in the cornea. J Clin Investig.
and motor neuron axons through chimeric peroneal 2016;126(5):1998–2011.
nerve grafts containing donor Schwann cells engi- 172. Ma MM, et  al. Involvement of ciliary neuro-
neered to express different neurotrophic factors. Exp trophic factor in early diabetic retinal neuropa-
Neurol. 2020;330:113355. thy in streptozotocin-induced diabetic rats. Eye.
155. Porzionato A, et al. Development of oxidized poly- 2018;32(9):1463–71.
vinyl alcohol-based nerve conduits coupled with 173. Saleh A, et  al. Ciliary neurotrophic factor acti-
the ciliary neurotrophic factor. Materials (Basel). vates NF-kappa B to enhance mitochondrial bio-
2019;12(12):1996. energetics and prevent neuropathy in sensory
156. Masu Y, et  al. Disruption of the Cntf gene neurons of streptozotocin-induced diabetic rodents.
results in motor-neuron degeneration. Nature. Neuropharmacology. 2013;65:65–73.
1993;365(6441):27–32. 174. Chowdhury SR, et  al. Ciliary neurotrophic fac-
157. Gatzinsky KP, et  al. Early onset of degenerative tor reverses aberrant mitochondrial bioenergetics
changes at nodes of Ranvier in alpha-motor axons of through the JAK/STAT pathway in cultured sensory
Cntf null (-/-) mutant mice. Glia. 2003;42(4):340–9. neurons derived from streptozotocin-induced dia-
158. Calcutt NA, et  al. Reduced ciliary neuronotro- betic rodents. Cell Mol Neurobiol. 2014;34(5):643–9.
phic factor-like activity in nerves from diabetic or 175. Lotz B, et al. A double-blind placebo-controlled clin-
galactose-­fed rats. Brain Res. 1992;575(2):320–4. ical trial of subcutaneous recombinant human ciliary
159. Mizisin AP, et  al. Aldose reductase inhibition neurotrophic factor (rHCNTF) in amyotrophic lat-
increases CNTF-like bioactivity and protein in sci- eral sclerosis. Neurology. 1996;46(5):1244–9.
atic nerves from galactose-fed and normal rats. 176. Ettinger MP, et  al. Recombinant variant of cili-
Diabetes. 1997;46(4):647–52. ary neurotrophic factor for weight loss in obese
160. Nakamura H, et  al. Olmesartan medoxomil ame- adults—a randomized, dose-ranging study. JAMA.
liorates sciatic nerve regeneration in diabetic rats. 2003;289(14):1826–32.
Neuroreport. 2009;20(16):1481–5. 177. Chew EY, et al. Effect of ciliary neurotrophic factor
161. Jiang Y, et al. Novel sites of aldose reductase immu- on retinal neurodegeneration in patients with macu-
nolocalization in normal and streptozotocin-diabetic lar telangiectasia type 2 a randomized clinical trial.
rats. J Peripher Nerv Syst. 2006;11(4):274–85. Ophthalmology. 2019;126(4):540–9.
150 N. A. Calcutt

178. Kauper K, et  al. Two-year intraocular delivery of 195. Yagihashi S.  Glucotoxic mechanisms and related
ciliary neurotrophic factor by encapsulated cell therapeutic approaches. Int Rev Neurobiol.
technology implants in patients with chronic retinal 2016;127:121–49.
degenerative diseases. Invest Ophthalmol Vis Sci. 196. Niimi N, et al. Aldose reductase and the polyol path-
2012;53(12):7484–91. way in Schwann cells: old and new problems. Int J
179. Rhode SC, Beier JP, Ruhl T.  Adipose tissue stem Mol Sci. 2021;22(3):1031.
cells in peripheral nerve regeneration–in vitro and 197. Tomlinson DR, Gardiner NJ.  Diabetic neuropa-
in vivo. J Neurosci Res. 2021;99(2):545–60. thies: components of etiology. J Peripher Nerv Syst.
180. Lara-Ramirez R, et  al. Expression of interleukin-6 2008;13(2):112–21.
receptor alpha in normal and injured rat sciatic 198. Jolivalt CG, et al. Defective insulin signaling path-
nerve. Neuroscience. 2008;152(3):601–8. way and increased glycogen synthase kinase-3
181. Rothaug M, Becker-Pauly C, Rose-John S. The role activity in the brain of diabetic mice: parallels with
of interleukin-6 signaling in nervous tissue. BBA-­ Alzheimer’s disease and correction by insulin. J
Mol Cell Res. 2016;1863(6):1218–27. Neurosci Res. 2008;86(15):3265–74.
182. Bolin LM, et  al. Interleukin-6 production by 199. King MR, et  al. Activation of the insulin-signaling
Schwann-cells and induction in sciatic-nerve injury. pathway in sciatic nerve and hippocampus of type 1
J Neurochem. 1995;64(2):850–8. diabetic rats. Neuroscience. 2015;303:220–8.
183. Gaudet AD, Popovich PG, Ramer MS.  Wallerian 200. Arvanitakis Z, et  al. Brain insulin signaling,
degeneration: gaining perspective on inflam- Alzheimer disease pathology, and cognitive func-
matory events after peripheral nerve injury. J tion. Ann Neurol. 2020;88(3):513–25.
Neuroinflammation. 2011;8:110. 201. Ferreira ST.  Brain insulin, insulin-like growth
184. Hirota H, et  al. Accelerated nerve regeneration factor 1 and glucagon-like peptide 1 signal-
in mice by upregulated expression of interleukin ling in Alzheimer’s disease. J Neuroendocrinol.
(IL) 6 and IL-6 receptor after trauma. J Exp Med. 2021;33(4):e12959.
1996;183(6):2627–34. 202. Rebelos E, et al. Brain glucose metabolism in health,
185. Andriambeloson E, et al. Interleukin-6 attenuates the obesity, and cognitive decline-does insulin have any-
development of experimental diabetes-related neu- thing to do with it? A narrative review. J Clin Med.
ropathy. Neuropathology. 2006;26(1):32–42. 2021;10(7):1532.
186. Cameron NE, Cotter MA. The neurocytokine, inter- 203. Sugimoto K, et al. Insulin receptor in rat peripheral
leukin-­6, corrects nerve dysfunction in experimental nerve: its localization and alternatively spliced iso-
diabetes. Exp Neurol. 2007;207(1):23–9. forms. Diabetes Metab Res Rev. 2000;16(5):354–63.
187. Cotter MA, et al. Effects of interleukin-6 treatment 204. Brussee V, Cunningham FA, Zochodne DW. Direct
on neurovascular function, nerve perfusion and vas- insulin signaling of neurons reverses diabetic neu-
cular endothelium in diabetic rats. Diabetes Obes ropathy. Diabetes. 2004;53(7):1824–30.
Metab. 2010;12(8):689–99. 205. Sugimoto K, Murakawa Y, Sima AA.  Expression
188. Cox AA, et al. Low-dose pulsatile interleukin-6 as a and localization of insulin receptor in rat dorsal
treatment option for diabetic peripheral neuropathy. root ganglion and spinal cord. J Peripher Nerv Syst.
Front Endocrinol. 2017;8:89. 2002;7(1):44–53.
189. Bishnoi M, et al. Streptozotocin-induced early ther- 206. Hackett AR, Strickland A, Milbrandt J.  Disrupting
mal hyperalgesia is independent of glycemic state insulin signaling in Schwann cells impairs myelin-
of rats: role of transient receptor potential vanilloid ation and induces a sensory neuropathy. Glia.
1 (TRPV1) and inflammatory mediators. Mol Pain. 2020;68(5):963–78.
2011;7:52. 207. Huang TJ, Verkhratsky A, Fernyhough P.  Insulin
190. Brini AT, et al. Therapeutic effect of human adipose-­ enhances mitochondrial inner membrane potential
derived stem cells and their secretome in experimen- and increases ATP levels through phosphoinosit-
tal diabetic pain. Sci Rep. 2017;7(1):9904. ide 3-kinase in adult sensory neurons. Mol Cell
191. Toledo-Corral CM, Banner LR.  Early changes of Neurosci. 2005;28(1):42–54.
LIFR and gp130 in sciatic nerve and muscle of dia- 208. Fernyhough P, et al. Insulin and insulin-like growth
betic mice. Acta Histochem. 2012;114(2):159–65. factor-I enhance regeneration in cultured adult-rat
192. Niemi JP, et al. Injury-induced gp130 cytokine sig- sensory neurons. Brain Res. 1993;607(1–2):117–24.
naling in peripheral ganglia is reduced in diabetes 209. Grote CW, et al. Deletion of the insulin receptor in
mellitus. Exp Neurol. 2017;296:1–15. sensory neurons increases pancreatic insulin levels.
193. Muona P, et  al. Glucose transporters of rat Exp Neurol. 2018;305:97–107.
peripheral-­nerve—differential expression of Glut1 210. Grote CW, Wright DE. A role for insulin in diabetic
gene by Schwann-cells and perineurial cells in vivo neuropathy. Front Neurosci. 2016;10:581.
and in vitro. Diabetes. 1992;41(12):1587–96. 211. Kobayashi M, Zochodne DW.  Diabetic neuropathy
194. Magnani P, et al. Glucose transporters in rat periph- and the sensory neuron: new aspects of pathogenesis
eral nerve: paranodal expression of GLUT1 and and their treatment implications. J Diabetes Investig.
GLUT3. Metabolism. 1996;45(12):1466–73. 2018;9(6):1239–54.
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 151

212. Kamiya H, et al. Unmyelinated fiber sensory neurop- 230. Lindfors L, et  al. Is GPR146 really the receptor
athy differs in type 1 and type 2 diabetes. Diabetes for proinsulin C-peptide? Bioorg Med Chem Lett.
Metab Res Rev. 2005;21(5):448–58. 2020;30(13):127208.
213. Pierson CR, et  al. Insulin deficiency rather than 231. Sima AAF, Kamiya H. Is C-peptide replacement the
hyperglycemia accounts for impaired neurotrophic missing link for successful treatment of neurological
responses and nerve fiber regeneration in type 1 complications in type 1 diabetes? Curr Drug Targets.
diabetic neuropathy. J Neuropathol Exp Neurol. 2008;9(1):37–46.
2003;62(3):260–71. 232. Wahren J, Kallas A, Sima AAF. The clinical poten-
214. Sima AAF.  The heterogeneity of diabetic neuropa- tial of C-peptide replacement in type 1 diabetes.
thy. Front Biosci. 2008;13:4808–16. Diabetes. 2012;61(4):761–72.
215. Stino AM, et  al. Evolving concepts on the role 233. Jolivalt CG, et  al. Efficacy of a long-acting
of dyslipidemia, bioenergetics, and inflamma- C-peptide analogue against peripheral neuropathy in
tion in the pathogenesis and treatment of dia- streptozotocin-diabetic mice. Diabetes Obes Metab.
betic peripheral neuropathy. J Peripher Nerv Syst. 2015;17(8):781–8.
2020;25(2):76–84. 234. Ekberg K, et al. Amelioration of sensory nerve dys-
216. Grote CW, et  al. Peripheral nervous system insu- function by C-peptide in patients with type 1 diabe-
lin resistance in ob/ob mice. Acta Neuropathol tes. Diabetes. 2003;52(2):536–41.
Commun. 2013;1:15. 235. Ekberg K, et al. C-peptide replacement therapy and
217. Grote CW, et  al. The role of insulin receptor sub- sensory nerve function in type 1 diabetic neuropathy.
strate 2  in neuronal insulin resistance in diabetic Diabetes Care. 2007;30(1):71–6.
neuropathy. J Peripher Nerv Syst. 2011;16:S49. 236. Johansson BL, et al. Beneficial effects of C-peptide
218. Aghanoori MR, et al. Insulin prevents aberrant mito- on incipient nephropathy and neuropathy in
chondrial phenotype in sensory neurons of type 1 patients with Type 1 diabetes mellitus. Diabet Med.
diabetic rats. Exp Neurol. 2017;297:148–57. 2000;17(3):181–9.
219. Chowdhury SKR, et  al. Mitochondrial respi- 237. Wahren J, et  al. Long-acting C-peptide and neu-
ratory chain dysfunction in dorsal root gan- ropathy in Type 1 diabetes: a 12-month clinical trial.
glia of streptozotocin-­ induced diabetic rats and Diabetes Care. 2016;39(4):596–602.
its correction by insulin treatment. Diabetes. 238. LeRoith D, Holly JMP, Forbes BE.  Insulin-like
2010;59(4):1082–91. growth factors: ligands, binding proteins, and recep-
220. Huang TJ, et al. Insulin prevents depolarization of the tors. Mol Metab. 2021;52:101245.
mitochondrial inner membrane in sensory neurons 239. Siddle K.  Molecular basis of signaling specificity
of type 1 diabetic rats in the presence of sustained of insulin and IGF receptors: neglected corners and
hyperglycemia. Diabetes. 2003;52(8):2129–36. recent advances. Front Endocrinol. 2012;3:34.
221. de la Hoz CL, et  al. A model of chronic diabetic 240. Gao WQ, et al. IGF-I deficient mice show reduced
polyneuropathy: benefits from intranasal insulin are peripheral nerve conduction velocities and decreased
modified by sex and RAGE deletion. Am J Physiol axonal diameters and respond to exogenous IGF-I
Endocrinol Metab. 2017;312(5):E407–19. treatment. J Neurobiol. 1999;39(1):142–52.
222. Francis GJ, et al. Motor end plate innervation loss in 241. Rabinovsky ED.  The multifunctional role of
diabetes and the role of insulin. J Neuropathol Exp IGF-1 in peripheral nerve regeneration. Neurol Res.
Neurol. 2011;70(5):323–39. 2004;26(2):204–10.
223. Guo GF, et al. Local insulin and the rapid regrowth 242. Ekstrom AR, Kanje M, Skottner A.  Nerve regen-
of diabetic epidermal axons. Neurobiol Dis. eration and serum levels of insulin-like growth
2011;43(2):414–21. factor-I in rats with streptozotocin-induced insulin
224. Singhal A, et  al. Near nerve local insulin prevents deficiency. Brain Res. 1989;496(1–2):141–7.
conduction slowing in experimental diabetes. Brain 243. Craner MJ, et al. Preferential expression of IGF-I in
Res. 1997;763(2):209–14. small DRG neurons and down-regulation following
225. Toth C, Brussee V, Zochodne DW.  Remote neuro- injury. Neuroreport. 2002;13(13):1649–52.
trophic support of epidermal nerve fibres in experi- 244. Kamiya H, et  al. C-Peptide reverses nocicep-
mental diabetes. Diabetologia. 2006;49(5):1081–8. tive neuropathy in type 1 diabetes. Diabetes.
226. Wahren J.  C-peptide and the pathophysiology of 2006;55(12):3581–7.
microvascular complications of diabetes. J Intern 245. Wuarin L, Guertin DM, Ishii DN.  Early reduction
Med. 2017;281(1):3–6. in insulin-like growth factor gene expression in dia-
227. Yosten GLC, Kolar GR. The physiology of proinsu- betic nerve. Exp Neurol. 1994;130(1):106–14.
lin C-peptide: unanswered questions and a proposed 246. Schmidt RE, et  al. Analysis of the Zucker
model. Physiology. 2015;30(4):327–32. Diabetic Fatty (ZDF) type 2 diabetic rat model
228. Hills CE, Brunskill NJ.  Cellular and physi- suggests a neurotrophic role for insulin/IGF-I
ological effects of C-peptide. Clin Sci. in diabetic autonomic neuropathy. Am J Pathol.
2009;116(7–8):565–74. 2003;163(1):21–8.
229. Yosten GLC, et  al. Evidence for an interaction 247. Zhuang HX, et  al. Insulin-like growth factor
between proinsulin C-peptide and GPR146. J (IGF) gene expression is reduced in neural tis-
Endocrinol. 2013;218(2):B1–8. sues and liver from rats with non-insulin-depen-
152 N. A. Calcutt

dent diabetes mellitus, and IGF treatment glycaemic control in adults with type 1 diabetes. Eur
ameliorates diabetic neuropathy. J Pharmacol Exp J Endocrinol. 2000;143(4):505–10.
Ther. 1997;283(1):366–74. 263. Tan K, Baxter RC. Serum insulin-like growth factor-
248. Busiguina S, et al. Neurodegeneration is associated ­I levels in adult diabetic-patients—the effect of age.
to changes in serum insulin-like growth factors. J Clin Endocrinol Metab. 1986;63(3):651–5.
Neurobiol Dis. 2000;7(6 Pt B):657–65. 264. Migdalis IN, et  al. Insulin-like growth-factor-I and
249. Han HJ, Kang CW, Park SH.  Tissue-specific regu- Igf-I receptors in diabetic-patients with neuropathy.
lation of insulin-like growth factors and insulin-like Diabet Med. 1995;12(9):823–7.
growth factor binding proteins in male diabetic rats 265. Crosby SR, et  al. Elevated plasma insulin-like
in  vivo and in  vitro. Clin Exp Pharmacol Physiol. growth factor binding protein-1 levels in type 1
2006;33(12):1172–9. (insulin-dependent) diabetic patients with peripheral
250. Hao W, et al. Hyperglycemia promotes Schwann cell neuropathy. Diabetologia. 1992;35(9):868–72.
de-differentiation and de-myelination via sorbitol 266. Simon CM, et  al. Dysregulated IGFBP5 expres-
accumulation and Igf1 protein down-regulation. J sion causes axon degeneration and motoneuron
Biol Chem. 2015;290(28):17106–15. loss in diabetic neuropathy. Acta Neuropathol.
251. Ishii DN, Lupien SB.  Insulin-like growth factors 2015;130(3):373–87.
protect against diabetic neuropathy: effects on sen- 267. Rabinovsky ED, Draghia-Akli R.  Insulin-like
sory nerve regeneration in rats. J Neurosci Res. growth factor I plasmid therapy promotes in  vivo
1995;40(1):138–44. angiogenesis. Mol Ther. 2004;9(1):46–55.
252. Lupien SB, Bluhm EJ, Ishii DN. Systemic insulin-­ 268. Saboory E, et  al. Exercise and insulin-like growth
like growth factor-I administration prevents cog- factor 1 supplementation improve angiogen-
nitive impairment in diabetic rats, and brain IGF esis and angiogenic cytokines in a rat model
regulates learning/memory in normal adult rats. J of diabetes-induced neuropathy. Exp Physiol.
Neurosci Res. 2003;74(4):512–23. 2020;105(5):783–92.
253. Schmidt RE, et  al. Insulin-like growth factor I 269. Aghanoori MR, et  al. Insulin-like growth fac-
reverses experimental diabetic autonomic neuropa- tor-1 activates AMPK to augment mitochondrial
thy. Am J Pathol. 1999;155(5):1651–60. function and correct neuronal metabolism in
254. Schmidt RE, et al. Effect of IGF-I and neurotrophin-3 sensory neurons in type 1 diabetes. Mol Metab.
on gracile neuroaxonal dystrophy in diabetic and 2019;20:149–65.
aging rats. Brain Res. 2000;876(1–2):88–94. 270. Aghanoori MR, et al. Sensory neurons derived from
255. Zhuang HX, et al. Insulin-like growth factors reverse diabetic rats exhibit deficits in functional glycolysis
or arrest diabetic neuropathy: effects on hyperalgesia and ATP that are ameliorated by IGF-1. Mol Metab.
and impaired nerve regeneration in rats. Exp Neurol. 2021;49:101191.
1996;140(2):198–205. 271. Wilson BD, et  al. Netrins promote develop-
256. Piriz J, Torres-Aleman I, Nunez A. Independent alter- mental and therapeutic angiogenesis. Science.
ations in the central and peripheral somatosensory 2006;313(5787):640–4.
pathways in rat diabetic neuropathy. Neuroscience. 272. Yam PT, Charron F.  Signaling mechanisms of
2009;160(2):402–11. non-conventional axon guidance cues: the Shh,
257. Wang H, et al. Protection of insulin-like growth fac- BMP and Wnt morphogens. Curr Opin Neurobiol.
tor 1 on experimental peripheral neuropathy in dia- 2013;23(6):965–73.
betic mice. Mol Med Rep. 2018;18(5):4577–86. 273. Anjaneyulu M, et  al. Transforming growth factor-­
258. Tang ZS, et  al. Peripheral pain is enhanced by beta induces cellular injury in experimental diabetic
insulin-like growth factor 1 and its receptors in a neuropathy. Exp Neurol. 2008;211(2):469–79.
mouse model of type 2 diabetes mellitus. J Diabetes. 274. Shimizu F, et  al. Advanced glycation end-products
2019;11(4):309–15. induce basement membrane hypertrophy in endo-
259. Li JB, et  al. The preventive efficacy of methylco- neurial microvessels and disrupt the blood-nerve
balamin on rat peripheral neuropathy influenced barrier by stimulating the release of TGF-beta and
by diabetes via neural IGF-1 levels. Nutr Neurosci. vascular endothelial growth factor (VEGF) by peri-
2010;13(2):79–86. cytes. Diabetologia. 2011;54(6):1517–26.
260. Song W, et al. Jinmaitong, a traditional Chinese com- 275. Resham K, et  al. Neuroprotective effects of iso-
pound prescription, ameliorates the streptozocin-­ quercitrin in diabetic neuropathy via Wnt/beta-­
induced diabetic peripheral neuropathy rats by catenin signaling pathway inhibition. Biofactors.
increasing sciatic nerve IGF-1 and IGF-1R expres- 2020;46(3):411–20.
sion. Front Pharmacol. 2019;10:255. 276. Resham K, Sharma SS. Pharmacologic inhibition of
261. Wang R, et  al. L-carnitine ameliorates peripheral porcupine, disheveled, and beta-catenin in Wnt sig-
neuropathy in diabetic mice with a corresponding naling pathway ameliorates diabetic peripheral neu-
increase in insulin-like growth factor-1 level. Mol ropathy in rats. J Pain. 2019;20(11):1338–52.
Med Rep. 2019;19(1):743–51. 277. Parmantier E, et  al. Schwann cell-derived desert
262. Ekman B, Nystrom F, Arnqvist HJ.  Circulating hedgehog controls the development of peripheral
IGF-I concentrations are low and not correlated to nerve sheaths. Neuron. 1999;23(4):713–24.
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 153

278. Moreau N, Boucher Y. Hedging against neuropathic 294. Taiana MM, et  al. Neutralization of Schwann cell-­
pain: role of hedgehog signaling in pathological secreted VEGF is protective to in vitro and in vivo
nerve healing. Int J Mol Sci. 2020;21(23):9115. experimental diabetic neuropathy. PLoS One.
279. Chapouly C, et  al. Impaired Hedgehog signalling-­ 2014;9(9):e108403.
induced endothelial dysfunction is sufficient 295. Chattopadhyay M, et al. HSV-mediated gene trans-
to induce neuropathy: implication in diabetes. fer of vascular endothelial growth factor to dorsal
Cardiovasc Res. 2016;109(2):217–27. root ganglia prevents diabetic neuropathy. Gene
280. Calcutt NA, et  al. Therapeutic efficacy of sonic Ther. 2005;12(18):1377–84.
hedgehog protein in experimental diabetic neuropa- 296. Murakami T, et al. VEGF 164 gene transfer by elec-
thy. J Clin Investig. 2003;111(4):507–14. troporation improves diabetic sensory neuropathy in
281. Stanton BZ, Peng LF.  Small-molecule modulators mice. J Gene Med. 2006;8(6):773–81.
of the Sonic Hedgehog signaling pathway. Mol 297. Schratzberger P, et  al. Reversal of experimental
Biosyst. 2010;6(1):44–54. diabetic neuropathy by VEGF gene transfer. J Clin
282. Leavitt E, Lask G, Martin S.  Sonic Hedgehog Investig. 2001;107(9):1083–92.
pathway inhibition in the treatment of advanced 298. Pawson EJ, et  al. Engineered zinc finger protein-­
basal cell carcinoma. Curr Treat Options Oncol. mediated VEGF-A activation restores deficient
2019;20(11):84. VEGF-A in sensory neurons in experimental diabe-
283. Li R, et  al. Heparin-poloxamer thermosensitive tes. Diabetes. 2010;59(2):509–18.
hydrogel loaded with bFGF and NGF enhances 299. Price SA, et al. Gene transfer of an engineered tran-
peripheral nerve regeneration in diabetic rats. scription factor promoting expression of VEGF-A
Biomaterials. 2018;168:24–37. protects against experimental diabetic neuropathy.
284. Nakae M, et al. Effects of bFGF on chronic neuropa- Diabetes. 2006;55(6):1847–54.
thy in STZ-diabetic rats. Diabetes. 2006;55:A188. 300. Hulse RP, et  al. Regulation of alternative VEGF-A
285. Foehring D, Brand-Saberi B, Theiss C.  VEGF-­ mRNA splicing is a therapeutic target for analgesia.
induced growth cone enhancement is diminished by Neurobiol Dis. 2014;71:245–59.
inhibiting tyrosine-residue 1214 of VEGFR-2. Cells 301. Hulse RP, et  al. Vascular endothelial growth fac-
Tissues Organs. 2012;196(3):195–205. tor-­
A(165)b prevents diabetic neuropathic pain
286. Sondell M, Sundler F, Kanje M. Vascular endothelial and sensory neuronal degeneration. Clin Sci.
growth factor is a neurotrophic factor which stimu- 2015;129(8):741–56.
lates axonal outgrowth through the flk-1 receptor. 302. Konishi Y, et  al. Trophic effect of erythropoietin
Eur J Neurosci. 2000;12(12):4243–54. and other hematopoietic factors on central cho-
287. Bates DO, et al. Physiological role of vascular endo- linergic neurons in  vitro and in  vivo. Brain Res.
thelial growth factors as homeostatic regulators. 1993;609(1–2):29–35.
Compr Physiol. 2018;8(3):955–79. 303. Campana WM, Misasi R, O’Brien JS. Identification
288. Arredondo-Garcia VK, et  al. association of the of a neurotrophic sequence in erythropoietin. Int J
vascular endothelial growth factor gene polymor- Mol Med. 1998;1(1):235–41.
phism+936 c/t with diabetic neuropathy in patients 304. Keswani SC, Leitz GJ, Hoke A.  Erythropoietin is
with type 2 diabetes mellitus. Arch Med Res. neuroprotective in models of HIV sensory neuropa-
2019;50(4):181–6. thy. Neurosci Lett. 2004;371(2–3):102–5.
289. Tavakkoly-Bazzaz J, et  al. VEGF gene polymor- 305. Collino M, et  al. Flipping the molecular switch
phism association with diabetic neuropathy. Mol for innate protection and repair of tissues: long-­
Biol Rep. 2010;37(7):3625–30. lasting effects of a non-erythropoietic small peptide
290. Zhang XH, et  al. Relationship between single engineered from erythropoietin. Pharmacol Ther.
nucleotide polymorphisms in the 3′-untranslated 2015;151:32–40.
region of the vascular endothelial growth fac- 306. Rey F, et  al. Erythropoietin as a neuroprotective
tor gene and susceptibility to diabetic periph- molecule: an overview of its therapeutic poten-
eral neuropathy in China. Arch Med Sci. tial in neurodegenerative diseases. ASN Neuro.
2014;10(5):1028–34. 2019;11:1759091419871420.
291. Chavez JC, Almhanna K, Berti-Mattera 307. Campana WM, Myers RR.  Erythropoietin and
LN. Transient expression of hypoxia-inducible fac- erythropoietin receptors in the peripheral ner-
tor-­1 alpha and target genes in peripheral nerves from vous system: changes after nerve injury. FASEB J.
diabetic rats. Neurosci Lett. 2005;374(3):179–82. 2001;15(8):1804–6.
292. Mo FF, et al. Prostaglandin E1 protects the periph- 308. Campana WM, Myers RR.  Exogenous erythropoi-
eral nerve in diabetics through preventing vascu- etin protects against dorsal root ganglion apoptosis
lar permeability changes. Exp Clin Endocrinol and pain following peripheral nerve injury. Eur J
Diabetes. 2018;126(2):113–22. Neurosci. 2003;18(6):1497–506.
293. Samii A, Unger J, Lange W.  Vascular endothelial 309. Li XQ, Gonias SL, Campana WM.  Schwann cells
growth factor expression in peripheral nerves and express erythropoietin receptor and represent a
dorsal root ganglia in diabetic neuropathy in rats. major target for Epo in peripheral nerve injury. Glia.
Neurosci Lett. 1999;262(3):159–62. 2005;51(4):254–65.
154 N. A. Calcutt

310. Bianchi R, et al. Erythropoietin both protects from 325. Pradat PF, et al. Continuous delivery of neurotrophin
and reverses experimental diabetic neuropathy. Proc 3 by gene therapy has a neuroprotective effect in
Natl Acad Sci U S A. 2004;101(3):823–8. experimental models of diabetic and acrylamide neu-
311. Demiot C, et al. Erythropoietin restores C-fiber func- ropathies. Hum Gene Ther. 2001;12(18):2237–49.
tion and prevents pressure ulcer formation in diabetic 326. Chu QM, et al. Systemic insulin-like growth factor-1
mice. J Invest Dermatol. 2011;131(11):2316–22. reverses hypoalgesia and improves mobility in a
312. Loesch A, et  al. Sciatic nerve of diabetic rat mouse model of diabetic peripheral neuropathy. Mol
treated with epoetin delta: effects on C-fibers and Ther. 2008;16(8):1400–8.
blood vessels including pericytes. Angiology. 327. Homs J, et  al. Intrathecal administration of IGF-I
2010;61(7):651–68. by AAVrh10 improves sensory and motor deficits
313. Chattopadhyay M, et  al. Neuroprotective effect in a mouse model of diabetic neuropathy. Mol Ther
of herpes simplex virus-mediated gene transfer of Methods Clin Dev. 2014;1:7.
erythropoietin in hyperglycemic dorsal root gan- 328. Ropper AH, et al. Vascular endothelial growth fac-
glion neurons. Brain. 2009;132:879–88. tor gene transfer for diabetic polyneuropathy: a
314. Wu Z, Mata M, Fink DJ.  Prolonged regulat- randomized, double-blinded trial. Ann Neurol.
able expression of EPO from an HSV vector 2009;65(4):386–93.
using the LAP2 promoter element. Gene Ther. 329. Eisenstein M.  Sangamo’s lead zinc-finger ther-
2012;19(11):1107–13. apy flops in diabetic neuropathy. Nat Biotechnol.
315. Brines M, et  al. Nonerythropoietic, tissue-­ 2012;30(2):121–3.
protective peptides derived from the tertiary struc- 330. Glassman AR, et al. Five-year outcomes after initial
ture of erythropoietin. Proc Natl Acad Sci U S A. aflibercept, bevacizumab, or ranibizumab treatment
2008;105(31):10925–30. for diabetic macular edema (protocol T extension
316. Leist M, et al. Derivatives of erythropoietin that are study). Ophthalmology. 2020;127(9):1201–10.
tissue protective but not erythropoietic. Science. 331. Striglia E, et  al. Emerging drugs for the treatment
2004;305(5681):239–42. of diabetic retinopathy. Expert Opin Emerg Drugs.
317. Bitto A, et al. Activation of the EPOR-beta common 2020;25(3):261–71.
receptor complex by cibinetide ameliorates impaired 332. Roy S, et al. Neurogenic tissue nanotransfection in
wound healing in mice with genetic diabetes. BBA-­ the management of cutaneous diabetic polyneuropa-
Mol Basis Dis. 2018;1864(2):632–9. thy. Nanomedicine. 2020;28:102220.
318. Schmidt RE, et  al. Effect of insulin and an 333. Obrien JS, et  al. Identification of prosaposin as
erythropoietin-­derived peptide (ARA290) on estab- a neurotrophic factor. Proc Natl Acad Sci U S A.
lished neuritic dystrophy and neuronopathy in Akita 1994;91(20):9593–6.
(Ins2(Akita)) diabetic mouse sympathetic ganglia. 334. Calcutt NA, et  al. Prosaposin gene expression and
Exp Neurol. 2011;232(2):126–35. the efficacy of a prosaposin-derived peptide in
319. Schmidt RE, et  al. Erythropoietin and its carba- preventing structural and functional disorders of
mylated derivative prevent the development of peripheral nerve in diabetic rats. J Neuropathol Exp
experimental diabetic autonomic neuropathy in Neurol. 1999;58(6):628–36.
STZ-induced diabetic NOD-SCID mice. Exp 335. Calcutt NA, Freshwater JD, O’Brien JS. Protection
Neurol. 2008;209(1):161–70. of sensory function and antihyperalgesic proper-
320. Brines M, et  al. ARA 290, a nonerythropoi- ties of a prosaposin-derived peptide in diabetic rats.
etic peptide engineered from erythropoietin, Anesthesiology. 2000;93(5):1271–8.
improves metabolic control and neuropathic symp- 336. Jolivalt CG, et al. Therapeutic efficacy of prosaposin-­
toms in patients with type 2 diabetes. Mol Med. derived peptide on different models of allodynia.
2014;20:658–66. Pain. 2006;121(1–2):14–21.
321. Zhang WJ, Yu GL, Zhang MY.  ARA 290 relieves 337. Jolivalt CG, et al. Impaired prosaposin secretion dur-
pathophysiological pain by targeting TRPV1 chan- ing nerve regeneration in diabetic rats and protection
nel: integration between immune system and noci- of nerve regeneration by a prosaposin-derived pep-
ception. Peptides. 2016;76:73–9. tide. J Neuropathol Exp Neurol. 2008;67(7):702–10.
322. Swartjes M, et al. ARA 290, a peptide derived from 338. King MR, et  al. Glycogen synthase kinase-3 inhi-
the tertiary structure of erythropoietin, produces bition prevents learning deficits in diabetic mice. J
long-term relief of neuropathic pain coupled with Neurosci Res. 2013;91(4):506–14.
suppression of the spinal microglia response. Mol 339. Mizisin AP, et  al. TX14(A), a prosaposin-derived
Pain. 2014;10:13. peptide, reverses established nerve disorders in
323. Mata M, Chattopadhyay M, Fink DJ. Gene therapy streptozotocin-diabetic rats and prevents them
for the treatment of sensory neuropathy. Expert Opin in galactose-fed rats. J Neuropathol Exp Neurol.
Biol Ther. 2006;6(5):499–507. 2001;60(10):953–60.
324. Chattopadhyay M, et  al. Prolonged preservation 340. Evans SR, et al. A randomized trial evaluating pro-
of nerve function in diabetic neuropathy in mice saptide (TM) for HIV-associated sensory neuropa-
by herpes simplex virus-mediated gene transfer. thies: use of an electronic diary to record neuropathic
Diabetologia. 2007;50(7):1550–8. pain. PLoS One. 2007;2(7):e551.
Neurotrophic Factors in the Pathogenesis and Treatment of Diabetic Neuropathy 155

341. Sung KJ, et  al. Swedish nerve growth fac- 351. Saleh A, et  al. Muscarinic toxin 7 signals via
tor mutation (NGF(R100W)) defines a role for Ca2+/calmodulin-dependent protein kinase
TrkA and p75(NTR) in nociception. J Neurosci. kinase beta to augment mitochondrial function
2018;38(14):3394–413. and prevent neurodegeneration. Mol Neurobiol.
342. Sung KJ, Yang WL, Wu CB.  Uncoupling neuro- 2020;57(6):2521–38.
trophic function from nociception of nerve growth 352. Calcutt NA, et  al. Selective antagonism of musca-
factor: what can be learned from a rare human dis- rinic receptors is neuroprotective in peripheral neu-
ease? Neural Regen Res. 2019;14(4):570–3. ropathy. J Clin Investig. 2017;127(2):608–22.
343. Yang WL, et al. Targeted mutation (R100W) of the 353. Jolivalt CG, et  al. Topical delivery of muscarinic
gene encoding NGF leads to deficits in the periph- receptor antagonists prevents and reverses peripheral
eral sensory nervous system. Front Aging Neurosci. neuropathy in female diabetic mice. J Pharmacol
2018;10:373. Exp Ther. 2020;374(1):44–51.
344. Yang WL, et al. A missense point mutation in nerve 354. Conese M, et  al. Harnessing stem cells and neuro-
growth factor (NGF(R100W)) results in selective trophic factors with novel technologies in the treat-
peripheral sensory neuropathy. Prog Neurobiol. ment of Parkinson’s disease. Curr Stem Cell Res
2020;194:101886. Ther. 2019;14(7):549–69.
345. Giuliani A, et  al. Effects of topical application of 355. Okawa T, et al. Transplantation of neural crest-like
CHF6467, a mutated form of human nerve growth cells derived from induced pluripotent stem cells
factor, on skin wound healing in diabetic mice. J improves diabetic polyneuropathy in mice. Cell
Pharmacol Exp Ther. 2020;375(2):317–31. Transplant. 2013;22(10):1767–83.
346. Hulse RP, et  al. The control of alternative splicing 356. Shibata T, et  al. Transplantation of bone marrow-­
by SRSF1 in myelinated afferents contributes to the derived mesenchymal stem cells improves
development of neuropathic pain. Neurobiol Dis. diabetic polyneuropathy in rats. Diabetes.
2016;96:186–200. 2008;57(11):3099–107.
347. Oltean S, et al. SRPK1 inhibition in vivo: modulation 357. Xie J, et  al. Therapeutic effects of stem cells from
of VEGF splicing and potential treatment for mul- human exfoliated deciduous teeth on diabetic periph-
tiple diseases. Biochem Soc Trans. 2012;40:831–5. eral neuropathy. Diabetol Metab Syndr. 2019;11:38.
348. Singh B, et  al. Regeneration of diabetic axons is 358. Hasegawa T, et al. Amelioration of diabetic periph-
enhanced by selective knockdown of the PTEN eral neuropathy by implantation of hematopoietic
gene. Brain. 2014;137:1051–67. mononuclear cells in streptozotocin-induced dia-
349. Fernyhough P, Calcutt N, Sabbir M.  Muscarinic betic rats. Exp Neurol. 2006;199(2):274–80.
receptor antagonism rescues cholinergic-mediated 359. Salgado AJ, et  al. Adipose tissue derived stem
cytoskeletal defects and enhances neurite out- cells secretome: soluble factors and their roles in
growth in sensory neurons. J Peripher Nerv Syst. regenerative medicine. Curr Stem Cell Res Ther.
2018;23(4):397–8. 2010;5(2):103–10.
350. Sabbir MG, Fernyhough P.  Muscarinic receptor 360. Yan H, Ding YF, Lu MJ. Current status and prospects
antagonists activate ERK-CREB signaling to aug- in the treatment of erectile dysfunction by adipose-­
ment neurite outgrowth of adult sensory neurons. derived stem cells in the diabetic animal model. Sex
Neuropharmacology. 2018;143:268–81. Med Rev. 2020;8(3):486–91.
Treatment Induced Neuropathy
of Diabetes

Nadia McMillan and Christopher H. Gibbons

1 Historical Background nosis also associated with diabetes mellitus) in


which patients develop profound weight loss, a
Treatment induced neuropathy of diabetes or symmetric peripheral neuropathy, and painful
TIND was coined in 2010 to describe the painful dysesthesias over the limbs and trunk without
somatosensory and autonomic changes following associated weakness [5]. The term acute painful
aggressive treatment of either type 1 or type 2 diabetic neuropathy encompasses these two
diabetes [1]. However, it was first observed by subtypes.
Caravati in 1933 [2]. He described a 48-year-old Since then, there have been countless case
female with diabetes started on insulin and reports describing the same changes in a handful
aggressive lifestyle management who developed of patients. In 2010, Gibbons and Freeman used
numbness, tingling, and shooting pains in her the term “Treatment induced neuropathy of dia-
lower extremities about 1 month after initiation betes” to describe a case series which described
of treatment. Within 3 days of discontinuing neuropathy in 16 patients with both type 1 and
insulin her symptoms improved and again type 2 diabetes [1]. In addition to the multiple
returned with re-initiation of insulin. Initial dis- case reports, they showed that autonomic symp-
cussion attributed these changes to an allergic toms correlated with the acute on set of neuropa-
skin reaction resulting in the term, “insulin neuri- thy associated with glycemic control in all
tis.” It was later described and termed diabetic patients and were able to provide longitudinal
neuropathic cachexia [3] followed by acute pain- follow up not previously performed.
ful diabetic neuropathy (APDN) by Archer et al. As it stands, TIND is now defined as “the
[4] It is important to point out that TIND is dis- acute onset of neuropathic pain and/or autonomic
tinct from diabetic neuropathic cachexia (a diag- dysfunction within 8 weeks of a large improve-
ment in glycemic control-specified as a decrease
in glycosylated hemoglobin A1C (HbA1c) of
N. McMillan ≥2% points over 3 months” [6]. Diagnostically,
Department of Neurology, Beth Israel Deaconess
Medical Center, Harvard Medical School,
the onset of symptoms can be useful when deter-
Boston, MA, USA mining the etiology of neuropathy. In a patient
C. H. Gibbons (*)
with diabetes the acute onset of neuropathy is
Beth Israel Deaconess Medical Center, likely to be related to TIND or a diabetic lumbo-
Boston, MA, USA sacral radiculoplexus neuropathy (DLRPN).
e-mail: cgibbons@bidmc.harvard.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 157
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_9
158 N. McMillan and C. H. Gibbons

Diabetic sixth nerve palsy would also fall into strength is typically preserved [9]. Motor symp-
this category. Diabetic lumbosacral radiculopa- toms are not considered a feature of TIND. Two
thy is an acute to subacute neuropathy in which other prominent features of TIND are autonomic
patients present with proximal or asymmetric symptoms and microvascular complications that
weakness, hypo or areflexia, and deep pain. There appear with or shortly after the onset of neuro-
is usually associated weight loss as well as wide- pathic pain [10]. Autonomic symptoms associ-
spread autonomic failure. Nerve biopsy in these ated with TIND include impotence, orthostatic
patients shows axonal loss and demyelination hypotension, syncope, hyperhidrosis, and anhi-
[7]. It is important to point out that both DLRPN drosis [1, 10, 11]. Microvascular complications
and TIND are distinctly separate from the nerve of TIND include both nephropathy and retinopa-
damage seen in diabetic polyneuropathy (DPN) thy [1, 10, 11].
in those with long standing diabetes [8]. Given The neuropathy associated with TIND affects
the well established presence of DPN, the topic small fibers including autonomic and somatosen-
of TIND itself has been controversial given sory with relative sparing of the large myelinated
­difficulty in teasing out pre-existing nerve dam- nerve fibers as determined by nerve conduction
age typically seen in long standing diabetes from studies and exam findings. Some patients present
that of an acute neuropathy. See below for clini- with prominent manifestations of autonomic dys-
cal features of TIND. function including orthostatic hypotension and
syncope. Gibbons et al. described 16 cases of adult
onset TIND in which all suffered autonomic dys-
2 Clinical Features function on testing and exhibited symptoms of
autonomic impairment. Autonomic testing showed
Clinical features of TIND include pain that is mild-to-moderate sympathetic and parasympa-
usually described as burning or shooting in a thetic dysfunction with strong correlations
length dependent, distal or diffuse, or proximal between the magnitude of decrease in HbA1c over
pattern that is frequently accompanied by allo- 3 months [10]. A greater change in HbA1c resulted
dynia and hyperalgesia. This usually occurs in worsening cardiovascular parasympathetic
within 2–8 weeks of initiation of aggressive function as determined by the heart rate response
treatment of lifestyle changes and correlates to deep respiration and the Valsalva maneuver and
with higher glucose and HbA1C scores. The tilt-table testing to 60° for 45  min. Sympathetic
larger and more rapid the correction, the greater adrenergic dysfunction was also observed and cor-
the neuropathy distribution and the more severe related with a greater change in HbA1c as deter-
the pain. Correction in HbA1c is often >2% in 3 mined by the fall in systolic blood pressure during
months. To further breakdown symptoms, using Valsalva and tilt-table testing [10].
a cohort of 16 patients with TINDs, Gibbons In patients with TIND there is a strong cor-
et  al. found that patients universally suffered relation with the simultaneous development of
from intractable pain that was typically described microvascular complications such as retinopa-
as symmetric and length dependent. However, thy and nephropathy. The retinopathy develops
3/16 of the patients with type 1 diabetes also rapidly in conjunction with the onset of pain [1,
described proximal/generalized pain. 10]. The Diabetes Control and Complications
Additionally, patients suffered from pain sensi- Trial (DCCT) defined retinopathy as the devel-
tivity with both hyperalgesia and allodynia opment of soft exudates and/or intraretinal
reported in 80% of type 1 subjects and 40% of microvascular abnormalities sand was consid-
type 2 patients. On neurologic examination, sen- ered “early” if it occurred between baseline and
sory changes typically include mild distal loss of 12-month follow-­up visits [12]. In their 2014
pain and vibratory senses in lower limbs, with study of 104 patients with TIND, Gibbons and
only rare absent ankle and knee reflexes, and Freeman determined that at baseline (prior to
normal reflexes in the upper extremities. Muscle TIND development) 65/104 did not have
Treatment Induced Neuropathy of Diabetes 159

r­etinopathy, 35/104 had non-­proliferative reti- of patients with TIND.  Gibbons and Freeman
nopathy, and 4/104 had proliferative retinopa- found that 87/104 patients with TIND had evi-
thy. One year after developing TIND, 10/104 dence of microalbuminuria 12 months after diag-
individuals had no retinopathy, 54/104 had non- nosis compared to 18/104 prior to development
proliferative retinopathy, and 40/104 had prolif- of TIND (P < 0.001, χ2). Those with stable glu-
erative retinopathy (P  <  0.001 vs. baseline cose did not have significant improvement at
values). Similar to the risk of developing TIND, 8-year follow-up and those with unstable glyce-
the risk of developing retinopathy increases mic control saw worsening in their renal function
with every percentage point decrease in the with some requiring initiation of hemodialysis
HbA1c over a 3-month period of time. The and renal transplantation [1, 6].
DCCT studies suggests that for every percent-
age point drop in HbA1C, there was a 1.6-fold
increase in the risk for early worsening retinop- 3 Pathophysiology
athy and correlates with the initial HbA1c level
[12, 13]. Notably, unstable glycemic control can Several theories exist regarding the underlying
be associated with severe proliferative retinopa- pathophysiology that leads to the development
thy resulting in vision loss [13] (Fig. 1). of TIND.  One hypothesis suggests that a ‘rela-
Nephropathy, as detected by microalbumin- tive’ hypoglycemia in those with chronically
uria on urine testing, is another prominent feature elevated blood glucose levels following ­treatment

Fig. 1  Treatment induced neuropathy of diabetes


160 N. McMillan and C. H. Gibbons

or aggressive lifestyles changes leads to an changes. Interestingly, neuropathy often occurs


energy dependent failure of axonal transport, in parallel with retinopathy and nephropathy sug-
apoptosis (cell death), and neuropathic pain as a gesting a common pathophysiological mecha-
consequence of axon regeneration and ectopic nism [1]. The possibility of a nutritional
firing of regenerating nerves [6, 14–16]. deficiency has been raised but is less likely given
Controlled studies of modest hypoglycemia in the absence of weight loss in patients who go on
humans using an insulin clamp have resulted in to develop TIND. Further clinical and molecular
the development of tactile hyperalgesia and tran- level studies need to be performed to evaluate the
sient autonomic dysfunction associated with the underlying mechanism.
release of pro-­inflammatory cytokines [17]. In
this theory, hypoglycemia acts as a stress
response resulting in the activation of the hypo- 4 Epidemiology
thalamic-pituitary adrenal axis and sympatho-
adrenal system and release of catecholamines, The true number of individuals at risk for TIND and
glucocorticoids, and cytokines in this disorder. the general prevalence in the population is unknown
In particular, cytokines such as interleukin-1β, likely because the majority of information has been
interleukin-6, and tumor necrosis factor-α have generated from case reports rather than prospective
been associated with painful neuropathy and are or longitudinal studies. Additionally, TIND remains
elevated in experimental hypoglycemia [10, 18, a relatively unrecognized complication of diabetes
19]. Using skin biopsies from eight patients with under reporting of symptoms [6, 20].
diagnosed with TIND, Gibbons and Freeman Prevalence estimates were initially assumed to
showed that there is diffuse damage to the unmy- be around 1%. However, more recent studies sug-
elinated and lightly myelinated nerve fibers that gest this number may be much higher. In a single
is temporally related to the rapid improvement in center study, Gibbons and Freeman found that
glucose control [1]. Further exploration of these 10.9% of patients referred to a tertiary care
models may provider greater understanding of ­diabetic neuropathy clinic over 5 years met crite-
the potential pathophysiological mechanisms of ria for diagnosis of TIND [6]. Potential epidemi-
the disease. ology can be extrapolated based on the DCCT
In another theorized mechanism, hyperglyce- trial that looked at early worsening retinopathy
mic induced microvascular changes including following initiation of treatment. In this trial,
arteriovenous shunts results in endoneurial isch- early worsening of retinopathy was seen in 22%
emia and firing of regenerating nerve fibers with of patient’s receiving intensive therapy vs. 13%
drops in blood glucose levels [15, 18, 19]. Here, in those assigned to conventional treatment [12,
the microvascular changes are unable to remodel 21–23]. Recovery was seen in 51% and 55% of
at the same rate of decline in serum glucose patients at their 18 month, respectively [12].
resulting in a sort of “steal syndrome” in which These findings suggest that TIND may be more
hyperproliferative vessels shunt blood away from common that previously suggested.
the endoneurium causing an acute neuropathy. A TIND has typically been reported in patient’s
role for microvascular factors has been supported newly diagnosed with type 2 diabetes and after
by the finding of abnormal epineurial vessel mor- initiation of insulin as well as those with poorly
phology on sural nerve photographs [15, 18, 19]. controlled type 1 diabetes who do not take their
In a cohort of five patient with TIND, Tesfaye medication appropriately. Typical age ranges are
found that all had arterial attenuation, arteriove- between 30 and 60 years at the time of diagnosis
nous shunts, and proliferating new vessels when [1]. While typically considered a diagnosis of
evaluated by sural nerve photography [15]. These adults, Alexandrou et  al. recently published a
proposed mechanisms suggest that hypoglyce- case series that included 7 children and young
mia or relative hypoglycemia from aggressive adults (age 9–22) diagnosed with TINDs [8].
management of diabetes results in neuro-vascular Prior to this only two case reports of TNDs in the
Treatment Induced Neuropathy of Diabetes 161

pediatric population had been reported [24, 25]. weight loss (>10 lb) developed TIND [26]. It is
In this case series, children with new onset or possible that diabetic anorexia creates a predis-
poorly controlled type 1 diabetes and elevated position to later nerve injury.
HbA1c (typically >14%) developed neuropathic
symptoms following initiation of treatment.
More specifically, 57% had new-onset diabetes 6 Complications
while the remainder had lived with it for 3–5
years. All presentations were complicated by There is a presumption that TIND tends to resolve
both autonomic symptoms (gastrointestinal with time with few long-term complications [14].
100%; cardiovascular 14%; genitourinary 14%) However, most patients experience months to
and microvascular complications (43% had years of severe neuropathic pain, symptoms of
microalbuminuria and 2/3 had non-proliferative autonomic dysfunction, and also develop compli-
retinopathy) [8]. This study was underpowered to cations of retinopathy and nephropathy. Long-
comment on epidemiology but, again, suggests term complications include a length dependent
that number of cases may be larger than expected small fiber neuropathy, renal failure, blindness,
due to underreporting. gastroparesis, orthostatic hypotension, and recur-
rent syncope [1, 6, 27].

5 Risk Factors
7 Long-Term Outcomes
TIND can develop in individuals with type 2 dia-
betes but is much more common among those The long-term outcome of neuropathic pain in
with T1DM. The main risk factors for TIND are TIND is generally good, but make take anywhere
rapid correction of serum glucose with either from 3 to 36 months before pain is under reason-
medication (insulin or oral hypoglycemic agent) able control without significant polypharmacy
or diet/lifestyle changes. Those with a higher [11, 26, 28]. In one study with the follow-up of 6
baseline HbA1c (>10% or 86 mmol/mol) are at diabetic patients with TIND for 2 years all
increased risk of TIND.  The absolute risk of reported only mild neuropathic pain in the feet
developing TIND was ~10% if the hemoglobin [28]. In contrast, in a larger longitudinal cohort
A1C (HbA1C) declined by more than 2% points study of 26 patients with TIND followed for 8
in 3 months. However, if the HbA1C declined by years, 19 of the individuals studied maintained
4% points or more in 3 months, the absolute risk stable glycemic control for the 8-year period of
of developing TIND exceeded 50% [11, 26]. A time. The other seven individuals had recurrent
large improvement in glycemic control >2% episodes of prolonged durations of hyperglyce-
points over 3 months is a risk factor and has been mia followed by periods of rapid glycemic con-
incorporated in the operational definition of trol and recurrence of pain, consistent with a
TIDN [6, 11]. No specific treatments for TIND diagnosis of recurrent TIND [11, 26]. Of those
have been studied [1, 6, 11]. with stable glycemic control, there was a gradual
Individuals with uncontrolled diabetes for improvement in neuropathic pain over 12–30
prolonged periods, particularly those with a his- months associated with either discontinuation of
tory of eating disorders involving insulin restric- decreased dosing of pain medication [27]. The
tion for calorie purging, termed diabetic anorexia, improvements were pathologically monitored
are at high risk. In a study by Gibbons and with longitudinal skin biopsies that confirmed an
Goeble-Fabri, 52/60 female and 10/16 males increase in intra-epidermal nerve fiber and
with T1DM had a history of an eating disorder improvement in autonomic function studies.
[26]. None of the 28 patients with type 2 diabetes There was also improvement in autonomic
and TIND had a history of eating disorders but 6 function testing and other microvascular parame-
individuals with type 2 diabetes with substantial ters such as a reduction in retinopathy and trend
162 N. McMillan and C. H. Gibbons

towards improving GFR over 8 years [1, 26]. In A physical representation of the risk of neu-
contrast, those seven individuals with unstable ropathy development is graphed by the change in
glycemic control in follow-up and severe progres- HbA1c. As the change in HbA1c increases
sion of neuropathy with motor involvement, renal (X-axis) the absolute risk of TIND development
failure, and severe proliferative retinopathy with increases (Y-axis) and is seen with the blue solid
blindness [27]. Despite the general improvement line. The physical distribution of neuropathy is
seen in the majority of patients with TIND over shown on the figures on the graph. The regions in
time, more than 25% of patients with TIND expe- red represent typical pain with neuropathy seen
rienced significant morbidity including amputa- in all patients, with regions in gray representing
tions, dialysis, and blindness [26]. Identification of pain in some patients. As the change in HbA1c
at-risk individuals is an unmet research priority. becomes larger, the risk of TIND increases, the
regional distribution of neuropathy increases and
the total pain scores increase as well.
8 Disease Management

The current management of TIND is primarily


symptomatic relief while simultaneously main-
References
taining glucose stability. Symptomatic therapy is 1. Gibbons CH, Freeman R. Treatment-induced diabetic
aimed at managing neuropathic pain which is neuropathy: a reversible painful autonomic neuropa-
often refractory to standard treatments. thy. Ann Neurol. 2010;67:534–41.
Neuropathic pain medications include the use of 2. Pomeranz B, Macaulay RJ, Caudill MA, et  al.
Assessment of autonomic function in humans
gabapentinoids, tricyclic antidepressants, or by heart rate spectral analysis. Am J Physiol.
serotonin-­norepinephrine reuptake inhibitors but 1985;248:H151–3.
there is no consensus on the management of 3. Ellenberg M.  Diabetic neuropathic cachexia.
TIND. Tricyclic antidepressants are often used in Diabetes. 1974;23:418–23.
4. Archer AG, Watkins PJ, Thomas PK, Sharma AK,
treatment of neuropathic pain, however, their use Payan J.  The natural history of acute painful neu-
in TIND can be limited due to their anticholinergic ropathy in diabetes mellitus. J Neurol Neurosurg
side effects that include orthostatic hypotension Psychiatry. 1983;46:491–9.
and syncope, which could exacerbate symptoms 5. Neal JM.  Diabetic neuropathic cachexia: a rare
manifestation of diabetic neuropathy. South Med J.
of autonomic dysfunction that is often seen in 2009;102:327–9.
TIND. Thus, neuropathic pain treatment is based 6. Gibbons CH, Freeman R. Treatment-induced neurop-
on the individual case and the associated symp- athy of diabetes: an acute, iatrogenic complication of
toms, but often requires polypharmacy for some diabetes. Brain. 2015;138:43–52.
7. Dyck PJ, Norell JE, Dyck PJ. Microvasculitis and
period of time for pain control to be optimized. ischemia in diabetic lumbosacral radiculoplexus neu-
It is not advised to allow for permissive hyper- ropathy. Neurology. 1999;53:2113–21.
glycemia to reduce neuropathic pain in TIND due 8. Alexandrou EG, Corathers SD, Lahoti A, et  al.
to the risk for longer term complications of recur- Treatment-induced neuropathy of diabetes in youth:
case series of a heterogeneous and challenging com-
rent TIND [26]. In general, a multi-disciplinary plication. J Endocr Soc. 2020;4(12):bvaa154.
approach that includes endocrine, neurology, and 9. Castellanos F, Mascias J, Zabala JA, Ricart C, Cabello
pain is beneficial. Finally, consideration should A, Garcia-Merino A. Acute painful diabetic neuropa-
also be given regarding prevention. By avoiding thy following severe weight loss. Muscle Nerve.
1996;19:463–7.
rapid and sizable changes to the HbA1c it may be 10. Gibbons CH, Freeman R. Treatment-induced neurop-
possible to prevent TIND development, although athy of diabetes: an acute, iatrogenic complication of
there is not data at this time to support this theory. diabetes. Brain. 2014;138:43–52.
While no clear consensus about rates of glycemic 11. Gibbons CH. Treatment induced neuropathy of diabe-
tes. Auton Neurosci. 2020;226:102668.
change exist, recommendations include a target 12. Group DR. Early worsening of diabetic retinopathy in
change in HbA1c of <2 percentage points over 3 the Diabetes Control and Complications Trial. Arch
months if feasible [6, 12, 13, 26]. Ophthalmol. 1998;116:874–86.
Treatment Induced Neuropathy of Diabetes 163

13. Lim SW, van Wijngaarden P, Harper CA, Al-Qureshi intensive glycaemic control. Clin Exp Ophthalmol.
SH.  Early worsening of diabetic retinopathy due 2019;47:265–73.
to intensive glycaemic control. Clin Experiment 22. Chantelau E, Meyer-Schwickerath R, Klabe
Ophthalmol. 2019;47:265–73. K.  Downregulation of serum IGF-1 for treatment
14. Knopp M, Srikantha M, Rajabally YA. Insulin neuri- of early worsening of diabetic retinopathy: a long-­
tis and diabetic cachectic neuropathy: a review. Curr term follow-up of two cases. Ophthalmologica.
Diabetes Rev. 2013;9:267–74. 2010;224:243–6.
15. Tesfaye S, Malik R, Harris N, et  al. Arterio-venous 23. Davis MD, Beck RW, Home PD, Sandow J, Ferris
shunting and proliferating new vessels in acute pain- FL. Early retinopathy progression in four randomized
ful neuropathy of rapid glycaemic control (insulin trials comparing insulin glargine and NPH insulin.
neuritis). Diabetologia. 1996;39:329–35. Exp Clin Endocrinol Diabetes. 2007;115:240–3.
16. Kihara M, Zollman PJ, Smithson IL, Lagerlund TD, 24. Chandler E, Brown M, Wintergerst K, Doll
Low PA.  Hypoxic effect of exogenous insulin on E. Treatment-induced neuropathy of diabetes (TIND)
normal and diabetic peripheral nerve. Am J Physiol. in pediatrics: a case report and review of the literature.
1994;266:E980–5. J Clin Endocrinol Metabol. 2019;105:395–8.
17. Gibbons CH, Adler GK, Bonyhay I, Freeman 25. Varadharaju N, Jeevarathnam D, Rajan M,
R. Experimental hypoglycemia is a human model of Ponnurangam Nagarajan V, James S.  A case of
stress-induced hyperalgesia. Pain. 2012;153:2204–9. treatment-induced neuropathy in an adolescent with
18. Llewelyn JG, Thomas PK, Fonseca V, King RHM, type 1 diabetes. Ann Pediatr Endocrinol Metab.
Dandona P. Acute painful diabetic neuropathy precip- 2019;24:203–6.
itated by strict glycaemic control. Acta Neuropathol. 26. Gibbons CH, Goebel-Fabbri A.  Microvascular
1986;72:157–63. complications associated with rapid improvements
19. Llewelyn JG, Thomas P, Fonseca V, Dandona in glycemic control in diabetes. Curr Diab Rep.
P. Acute painful diabetic neuropathy. Diabetes Care. 2017;17:48.
1988;11:748–9. 27. Gibbons CH.  Treatment induced neuropathy of
20. Nicodemus JM, Enriquez C, Marquez A, Anaya diabetes-­long term implications in type 1 diabetes. J
CJ, Jolivalt CG.  Murine model and mechanisms Diabetes Complications. 2017;31(4):715–20.
of treatment-induced painful diabetic neuropathy. 28. Dabby R, Sadeh M, Lampl Y, Gilad R, Watemberg
Neuroscience. 2017;354:136–45. N.  Acute painful neuropathy induced by rapid cor-
21. Lim SW, van Wijngaarden P, Harper CA, Al-Qureshi rection of serum glucose levels in diabetic patients.
SH.  Early worsening of diabetic retinopathy due to Biomed Pharmacother. 2009;63:707–9.
Asymmetric Diabetic Neuropathy:
Radiculoplexus Neuropathies,
Mononeuropathies, and Cranial
Neuropathies

Pariwat Thaisetthawatkul and P. James B. Dyck

1 Introduction neuropathies [2] (Table  1). Asymmetric dia-


betic neuropathies consist of three major types:
Diabetic neuropathy is the most common diabetic radiculoplexus neuropathies, diabetic
acquired neuropathy in the world. It can be mononeuropathies, and diabetic cranial neu-
seen in up to 50% in diabetic patients [1]. The ropathies [2]. Even though asymmetric dia-
clinical manifestations of diabetic neuropathy betic neuropathies are less common than
vary, ranging from predominantly sensory symmetric diabetic neuropathies, they are still
symptoms sometimes with associated neuro- important to correctly diagnose as they have
pathic pain in diabetic sensorimotor polyneu- different underlying pathophysiology, may be
ropathy to asymmetrical or multifocal misdiagnosed as the other forms of neuropa-
weakness, pain, and sensory loss in diabetic thies with similar clinical manifestations and
radiculoplexus neuropathy. There are different perhaps most importantly may respond to
ways of classifying diabetic neuropathy. One immunotherapy or other therapeutic interven-
way is dividing diabetic neuropathy into two tions. This chapter describes the asymmetric
major categories: symmetric and asymmetric diabetic neuropathies in details.

P. Thaisetthawatkul
Department of Neurological Sciences, University of
Nebraska Medical Center, 988435 Nebraska Medical
Center, Omaha, NE, USA
e-mail: pthaiset@unmc.edu
P. J. B. Dyck (*)
Department of Neurology, Mayo Clinic College of
Medicine, Rochester, MN, USA
e-mail: Dyck.PJames@mayo.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 165
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_10
166 P. Thaisetthawatkul and P. J. B. Dyck

Table 1  Classification of diabetic neuropathies


Symmetrical neuropathies Asymmetrical neuropathies
1. Categories 1. Categories
Diabetic distal sensorimotor neuropathy (DPN) Diabetic radiculoplexus neuropathy (DRPN)
Diabetic autonomic neuropathy (DAN)  –  Diabetic lumbosacral radiculoplexus neuropathy
Diabetic small fiber neuropathy (DLRPN)
Treatment-induced neuropathy (TIN)  –  Diabetic cervical radiculoplexus neuropathy (DCRPN)
 –  Diabetic thoracic radiculoneuropathy (DTRN)
Diabetic mononeuropathies
 –  Median mononeuropathy across the wrist (MNW, carpal
tunnel syndrome)
 –  Ulnar mononeuropathy across the elbow (UNE)
 –  Fibular mononeuropathy (FN)
 –  Lateral femoral cutaneous mononeuropathy (meralgia
paresthetica (MP))
Diabetic cranial neuropathies
 –  Diabetic oculomotor mononeuropathies (OMM)
 –  Bell’s palsy
2. Pathophysiology 2. Pathophysiology
Metabolic derangement Inflammatory microvasculitis
Altered microvessel functions Ischemic nerve injury
Increased oxidative stress Compression
3. Clinical presentation 3. Clinical presentation
Symmetrical distal Asymmetrical distal and/or proximal
Predominantly sensory or autonomic symptoms Motor greater than sensory or autonomic involvement

upper limbs or truncal area separately or in com-


2 Diabetic Radiculoplexus bination and having a combination of DLRPN,
Neuropathies (DRPN) DCRPN, and DTRN occurring in one setting has
been described in about one-third of cases [5, 6].
DRPN are clinical syndromes usually described
as acute to subacute onset of pain followed by
weakness and sensory symptoms starting asym- 3 Diabetic Lumbosacral
metrically in one limb and usually spread to the Radiculoplexus Neuropathy
other corresponding limb in weeks or a few (DLRPN)
months in diabetic patients [3]. Anatomically,
DRPN clinically and pathologically involve the 3.1 Historical Perspective
roots, plexus, and peripheral nerves (hence the
terminology of radiculoplexus neuropathy). DLRPN is a long recognized asymmetric painful
DRPN comes in three types that occur in isolation paralytic lower limb syndrome of acute to subacute
or present together. If DRPN affects predomi- onset in diabetic patients that has been called by
nantly the lower limbs, it is called diabetic lumbo- many different names including diabetic myelopa-
sacral radiculoplexus neuropathy (DLRPN) while thy [7], diabetic amyotrophy [8], diabetic femoral-
predominant upper limb involvement gives a clin- sciatic neuropathy [9], diabetic femoral neuropathy
ical syndrome of diabetic cervical radiculoplexus [10], diabetic mononeuropathy multiplex [11],
neuropathy (DCRPN) [3]. In a case of truncal proximal diabetic neuropathy [12, 13], Bruns-
involvement, the term diabetic thoracic radiculo- Garland syndrome [14, 15], diabetic polyradicu-
neuropathy (DTRN) is used to describe the clini- lopathy [16], painful lumbosacral plexopathy [17],
cal syndrome [4]. Among all these subtypes, diabetic chronic inflammatory demyelinating poly-
DLRPN is probably the most prevalent [4]. DRPN radiculoneuropathy [18], multifocal diabetic neu-
may occur in isolation affecting the lower limbs, ropathy [19], and diabetic lumbosacral
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 167

radiculoplexus neuropathy [5, 20]. These different common form of inflammatory neuropathy. It is
names were given to DLRPN based on different clear from this study that the incidence of DLRPN
suspected regions of anatomical involvement (spi- is higher than that of other commonly found
nal cord, muscles or peripheral nerves) or based on inflammatory polyneuropathies such as Guillain-­
different purported pathophysiological mechanisms Barre syndrome or chronic inflammatory demy-
(metabolic derangement, ischemia of nerve from elinating polyradiculoneuropathy (CIDP).
non-inflammatory causes or from inflammatory Another important conclusion from this study is
causes) documented among many studies [7–19]. that diabetes mellitus is clearly a risk factor for
These studies emphasized different types of clinical the development of LRPN as it occurred eight
presentations, different anatomical involvements, times more frequently in diabetic persons [22].
and different biopsy or autopsy findings [7–19]. A
large prospective clinical and pathological study by
Dyck et al. in 33 DLRPN patients compared to dia- 3.3 Clinical Presentation
betic polyneuropathy patients showed strong evi-
dence to support that ischemic nerve injury from Typically, DLRPN usually starts with the heralding
microvasculitis is the main underlying pathophysi- symptom of focal pain, often severe and sharp,
ological mechanism of DLRPN [5]. shooting or burning in quality and associated with
contact allodynia (normal touch producing a pain-
ful experience), followed by weakness that usually
3.2 Epidemiology starts proximally more commonly than distally
usually in one lower limb. Over weeks to months,
As mentioned above, the descriptions of DLRPN the pain and weakness spreads to the initially unaf-
were based on case reports or care series and the fected segments and then often to the other lower
incidence of DLRPN has been unknown due to limb [23]. The onset often occurs in the setting of a
lack of population-based epidemiological stud- large amount of weight loss and often after altera-
ies. Furthermore, experts of DLRPN have tions in the diabetic medications so that by the time
assumed that it is associated with diabetes melli- of evaluation the blood sugar is frequently under
tus; however, a non-diabetic form of LRPN has good control [5]. When a DLRPN patient is seen
been identified; it had never been shown that the by a neurologist, the patient usually has problem-
rate of diabetes mellitus is increased in LRPN atic motor deficits, often sustaining falls and many
[21]. Recently, the incidence of DLRPN was are not able to walk and require use of a wheelchair
evaluated in a population-based epidemiological [23]. The pain of DLRPN usually subsides before
study conducted in a northern United States com- the weakness improves. The clinical course of
munity by Ng et al. [22] The study was done in DLRPN is that of a monophasic illness with maxi-
the well-defined population of Olmsted County mum motor deficits and disease nadir occurring on
MN and the study assessed the incidence of average at around 6 months, followed by gradual
LRPN (both diabetic and non-diabetic) and the improvement occurring over many months (and
relationship of LRPN with diabetes mellitus [22]. sometimes years) [5]. Many patients do not have
The overall incidence of LRPN was found to be complete recovery and proximal segments reinner-
4.13/100,000/year (95% CI 3.12–5.15) with the vated more completely than distal segments result-
incidence of DLRPN was 2.57/100,000/year ing in many patients being left with a footdrop [5].
(95% CI 1.76–3.38) and non-DLRPN was Autonomic symptoms such as orthostatic hypoten-
1.6/100,000/year (95% CI 0.97–2.24) [22]. The sion, urinary bladder dysfunction, gastrointestinal
odds of having LRPN among diabetes patients symptoms, and tachycardia can be seen in about
was 7.91 (95% CI 4.11–15.21) but the odds of half of the DLRPN patients [5]. These clinical fea-
having LRPN among pre-diabetes patients was tures of DLRPN distinguish it from a more com-
1.006 (95% CI 1.00–1.01) [22]. An important mon diabetic sensorimotor polyneuropathy as
conclusion of this study is that LRPN is a very diabetic polyneuropathy has a more insidious
168 P. Thaisetthawatkul and P. J. B. Dyck

onset, more sensory predominant symptoms infre- that has little associated weakness but with electro-
quent distal motor weakness without proximal physiological studies showing motor involvement
weakness, more symmetrical presentation and usu- that met the DLRPN criteria (involvement in at
ally occurring in patients with a prolonged history least two peripheral nerves from at least two nerve
of diabetes mellitus or with poor blood glucose root distributions) [25]. It is likely that most of
control [23]. The spectrum of DLRPN also extends these sensory dominant cases are not correctly
to a more uncommon clinical presentation of a diagnosed as being DLRPN.
painless motor predominant, diabetic neuropathy
(PMPDN) [24]. PMPDN has slower onset, more
symmetrical findings and more severe motor weak- 3.4 Pathophysiology
ness, more associated upper limb involvement, and
no pain symptoms compared to typical DLRPN DLRPN is caused by ischemic nerve injury asso-
cases which has more patchy distribution and is ciated with increased nerve inflammation and
multifocal [24]. In an epidemiological study, Pinto microvasculitis [5, 17, 19, 25]. Nerve biopsies in
and colleagues noted that a sensory predominant DLRPN usually show findings of ischemic nerve
form of DLRPN occasionally exists in which pain injury including multifocal fiber loss (Fig.  1d),
and sensory loss of the lower limb are prominent focal perineurial degeneration and thickening,

a b

c d

Fig. 1  Serial paraffin sections (a–c) and epoxy semithin stain) causing disruption of muscle layer (c: smooth mus-
section (d) from a sural nerve biopsy in a patient with dia- cle actin [SMACTIN] stain) and multifocal nerve fiber
betic lumbosacral radiculoplexus neuropathy (DLRPN) loss within a fascicle (d: methylene blue stain). These sec-
shows epineurial perivascular inflammatory cell infiltrates tions show finding of microvasculitis and ischemic nerve
that involve the microvessel wall (a: H&E stain; b: CD 45 injury as are typically seen in DLRPN
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 169

injury neuroma with abortive regeneration of plexopathy counts as two roots levels) with lum-
axons, neovascularization, and increased rate of bosacral paraspinal denervation being allowed.
axonal degeneration and secondary segmental These electrophysiological findings are required
demyelination (grouped areas of demyelination to make a definite diagnosis and at the same time
clustered along one teased nerve fiber due to axo- exclude lumbosacral radiculopathy or mononeu-
nal dystrophy) [5]. Perivascular inflammatory ropathy which can have similar presentations to
infiltrates of varying sizes surrounding and DLRPN [23]. A sensory nerve biopsy from the
involving epineurial (Fig. 1a, b) more than endo- lower limb is not required for the diagnosis pro-
neurial blood microvessels are usually present vided the clinical presentation is typical.
[5]. In some cases, there is infiltration and However, when the clinical presentation is atypi-
destruction of the vessel wall by the inflamma- cal or the disease course is prolonged and it is
tory infiltrates suggesting microvasculitis; this unclear if the inflammatory neuropathy is still
can be confirmed with smooth muscle actin active, a sensory nerve biopsy may provide histo-
(SMACTIN) staining (Fig.  1c) of these blood logic findings to support the diagnosis. The dif-
vessels showing distortion of the architecture of ferential diagnosis of DLRPN include neoplastic
these blood vessels consistent with microvasculi- lumbosacral plexopathy from adjacent pelvic
tis [5]. Evidence of prior bleeding into the nerve organs such as colon, cervix, ovaries, and urinary
tissue is seen in two thirds of cases as evidenced bladder or distant metastasis from lungs, breast
by the presence of hemosiderin-laden macro- and lymphoma [3], infection such as perirectal
phages in the perineurium and the epineurium abscess [28], tuberculosis [29] or diffuse infiltra-
often adjacent to damaged microvessels [5]. tive lymphomatosis syndrome in HIV patients
These biopsy findings are similar between [30], sarcoidosis, traumatic lumbosacral plexopa-
DLRPN and non-diabetic LRPN with both sug- thy [3], radiation-induced lumbosacral plexopa-
gestive of an inflammatory ischemic pathology thy [3], hematoma and vascular lesions in the
which is evidence that there is a common patho- pelvis [3], and postsurgical inflammatory neu-
physiology between the two clinical entities [21, ropathy [31]. It is therefore important to obtain a
26]. PMPDN also has similar nerve biopsy find- detailed and meticulous history and appropriate
ings to DLRPN providing evidence that PMPDN laboratory tests such as blood tests, a spinal tap
is in fact a variant of DLRPN and not diabetic and appropriate neuroimaging studies to exclude
CIDP [24]. Upregulation of inflammatory media- the other conditions.
tors such as intercellular adhesion molecule-1,
tumor necrosis factor-α, interleukin-6, and
nuclear factor ƙB have been demonstrated in the 3.6 Management
nerve tissue of patients with DLRPN and non-­
DLRPN supporting the role of inflammation in As the pathophysiology of DLRPN is ischemic
the pathophysiology of DLRPN [27]. nerve injury related to inflammatory microvascu-
litis, the mainstay of treatment for DLRPN is
immunotherapy. As there are no controlled clini-
3.5 Diagnosis cal studies that unequivocally show the efficacy
of immunotherapy in DLRPN, the use of immu-
The diagnosis of DLRPN requires a typical clini- notherapy is considered a good practice based on
cal presentation (as described above), compatible the pathophysiology and clinical experience [23].
abnormalities in electrophysiologic studies, and In a blinded placebo-controlled study of intrave-
the other causes excluded. The nerve conduction nous methylprednisolone (IVMP) given intermit-
study and electromyography (NCS/EMG) crite- tently over 3  months (pulsed), no significant
ria for DLRPN are neurogenic involvement in differences in time to improvement by 4 points of
distribution of at least two peripheral nerves from the Neuropathy Impairment Score occurred
at least two lumbosacral roots (upper lumbar between the treated and the placebo DLRPN
170 P. Thaisetthawatkul and P. J. B. Dyck

groups were found [32]. However, patient’s per- lem is many patients. Data from the Olmsted
ception of pain and positive sensory symptoms County epidemiological DLRPN study suggests
were significantly improved in the methylpred- that community-based DLRPN is less severe than
nisolone group compared to placebo [32]. In referral-based DLRPN; fewer community
another study of open trial of pulsed IVMP in DLRPN patients are in wheelchairs or have bilat-
non-diabetic LRPN, there was significant eral disease (37% community compared to 92%
improvement in neurologic examination in all referral) but that the overall features of the
patients (but the authors note that LRPN is a community-­based illness are similar to the pub-
monophasic illness so spontaneous improvement lished series (the illness begins focally with pain,
is expected) [33]. In an open trial of pulsed oral followed by weakness and begins in proximal or
and intravenous methylprednisolone in DLRPN distal segments but usually eventually involves
patients, improvement of the neuropathy was both) [25]. The long-term survival is reduced in
noted [34]. The doses, routes, and types of corti- DLRPN compared to community control patients
costeroid used in DLRPN vary in clinical but that this reduction in survival is secondary to
­practices [35]. In our practice, we generally use a diabetes mellitus and its co-morbidities rather
dose of 1 g IVMP given three times in the first than the neuropathy itself [25].
week (usually every other day) followed by 1 g
IVMP weekly over 12 weeks. Intravenous immu-
noglobulin (IVIG) or plasma pheresis may be 4 Diabetic Cervical
considered but have fewer studies to support their Radiculoplexus Neuropathy
efficacy in DLRPN [36]. It is important to moni- (DCRPN)
tor for and manage possible side effects from cor-
ticosteroid including hyperglycemia (particularly In 1890, Auche reported 11 cases with different
the use in diabetic patients), weight gain, edema, types of diabetic peripheral neuropathies and
bone necrosis, osteoporosis, glaucoma, cataract, among these described three cases of focal lower
infections, steroid-induced psychosis, sleep dis- (probable DLRPN) and two cases of focal upper
turbance, and mood swing. As severe neuropathic limb (probable DCRPN) pain and weakness [37].
pain and significant motor weakness may develop Other cases of CRPN were described in 1948 as
in this condition, pain management, physical the shoulder-girdle syndrome or neuralgic amy-
therapy, and rehabilitation must be considered otrophy [38]. The term brachial plexus neuropa-
important management strategies for this group thy was later adopted for this condition of rapid
of patients. onset of pain followed by weakness and sensory
loss in the upper limbs [39]. Most series of CRPN
did not record the occurrence of diabetes mellitus
3.7 Outcome and Prognosis among those affected [38, 40] but in a large study
of 99 CRPN patients, there were five patients
As DLRPN is usually monophasic illness with who also had diabetes mellitus [39]. The inci-
self-improvement, most patients’ disease course dence of DCRPN in the general population is
will plateau after months of illness and then they unknown. In a large cohort of longitudinal
will start to recover. However, many patients do population-­based study of diabetic neuropathy,
not experience a complete recovery and some the frequency of DCRPN is estimated to be less
degree of residual deficits are common [5]. In a than 1% [1]. DCRPN is a distinct clinical syn-
large prospective study on DLRPN patients, at drome of acute or subacute progressive pain,
2-year follow-up, 36% of the patients were able weakness, and sensory symptoms that usually
to walk independently but 9% continued to start in one upper extremity and spreads to the
require a wheelchair and 48% continued to use other segment of the same upper limb or to the
walking aids [5]. Neuropathic pain and long-term other upper limb in an asymmetrical manner
morbidity from weakness continue to be a prob- among diabetic patients [3]. In a large cohort of
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 171

DCRPN seen at one institution, DCRPN usually management of DCRPN is also similar to the
starts acutely and unilaterally in one upper limb management of DLRPN [6]. Immunotherapy
and spreads to the other upper limb (similar to such as corticosteroid, IVIg or plasmapheresis
how DLRPN becomes bilateral) [6]. Involvement have been used in DCRPN and variable improve-
of the other body regions producing co-occurring ment has been reported in this condition even
DLRPN, DTRN or phrenic neuropathy was also though treated patients had more severe disability
common [6]. Motor weakness is the main cause than untreated patients [6]. There has been no
of disability in DCRPN and it usually reaches its clinical trial of immunotherapy in DCRPN.
maximum severity more quickly than does the
deficits in DLRPN (often within 1 week after the
onset) [6]. Similar to DLRPN, weight loss may 5 Diabetic Thoracic
be seen at the onset of the symptoms but less Radiculoneuropathy (DTRN)
common than DLRPN [6]. Accompanying auto-
nomic symptoms such as orthostatic lightheaded- DTRN is a clinical syndrome of acute–subacute
ness and changes in sweating can be seen in onset of pain, often described as burning, sharp
about 30% [6]. DCRPN is diagnosed based on or shooting, jabbing and throbbing characters,
compatible symptoms and signs and abnormali- affecting the thoracic cage, upper abdomen or
ties in electrophysiologic studies indicating bra- scapula areas in a radicular pattern [41]. An early
chial plexus involvement [6]. The description of DTRN in diabetic patients was dia-
electrophysiologic abnormalities usually are betic truncal mononeuropathy [42]. The pain
more widespread than the clinical severity [6]. usually started in an acute, usually rapid, or sub-
The other causes of CRPN needs to be excluded acute manner and could last for many months or
by appropriate blood and CSF tests as well as more than a year before a patient seeks medical
neuroimaging studies [3]. Serological markers of attention [41]. It can be unilateral or bilateral but
a more widespread inflammatory response such asymmetrical and can involve several dermato-
as sedimentation rate and markers of autoimmu- mal segments [41]. It can affect mainly the
nity such as anti-nuclear antibodies are usually abdominal area giving a false impression of acute
elevated [6]. The pathophysiology of DCRPN abdomen leading patients to wrongly undergo an
has been reported in a large cohort of DCRPN unnecessarily exploratory laparotomy [43].
patients to show ischemic nerve injury and Unilateral abdominal muscle weakness causing
immune-mediated microvasculitis similar to hernia or protrusion of the abdominal wall has
what was reported in DLRPN [6]. Nerve biopsies been described [44]. In many cases, the patients
in DCRPN shows multifocal fiber loss, focal give a history of paresthesia, sensory loss or con-
perineurial degeneration and thickening and tact allodynia at the thoracic cage or abdominal
injury neuroma suggesting ischemic nerve injury wall suggestive of the diagnosis. In general, the
similar to the pathology of DLRPN [6]. Interstitial duration of diabetes mellitus in DTRN cases is of
abnormalities also include perivascular inflam- long duration, with a mean duration of 12 years
matory infiltrates involving vessel walls, vessel in one study [41]. Similar to DLRPN and
wall disruption, hemosiderin in macrophages, DCRPN, accompanying weight loss at the time
bleeding into the nerve interstitial tissue, and of onset can be seen up to 60% of patients [41].
neovascularization that were in some cases diag- Cachexia at the onset is another common com-
nostic of microvasculitis similar to DLRPN [6]. plaint in DTRN [45]. The diagnosis of DTRN
Similar pathologic changes suggest that DCRPN requires the recognition of the entity as the symp-
and DLRPN are both subtypes of the broader toms may mimic the other causes of thoracic
diagnosis of DRPN. A nerve biopsy is not always radiculopathy, most commonly degenerative tho-
indicated to make a diagnosis but may be needed racic disc disease [45] and herpes zoster [46].
in a DCRPN case with atypical clinical presenta- The other causes of thoracic radicular pain
tion. Because of similar pathophysiology, the include Lyme neuroborreliosis [47], neurosar-
172 P. Thaisetthawatkul and P. J. B. Dyck

coidosis [48], benign [49], and malignant [50, be explained by several reasons. One morpho-
51] mass lesions and myodil cyst after having a metric study of posterior interosseous nerve in
myelogram [52]. It is therefore important to get a diabetic patients who have CTS showed signifi-
detailed history and physical examination and cantly increased nerve vessel area, basement
appropriate tests with neuroimaging study to membrane area, and density of unmyelinated
exclude the other causes. Confirmation of DTRN axons and expression of vascular endothelial
requires the findings of fibrillation potentials or growth factor indicating nerve ischemia and
positive waves at the rectus abdominis muscles endoneurial hypoxia enhancing increased vascu-
and in the thoracic paraspinal muscles [45, 53]. lar permeability and neovascularization in dia-
DTRN may coexist with DCRPN or DLRPN in betic CTS compared to non-diabetic CTS patients
many cases [5, 6]. The course and prognosis of while density of myelinated fibers, myelinated
DTRN is that of self-limited symptoms in several fiber area, and axon area are significantly
months [41]. The main treatment of DTRN is decreased [57]. In addition, ultrasonographic
pain management. In most cases, there is not an studies in peripheral nerves in diabetic patients
indication for immunotherapy but the authors show diffuse enlargement of nerve cross-­sectional
have used weekly pulse intravenous methylpred- areas at non-entrapment sites in diabetic patients
nisolone for 6–12 weeks in some cases with sat- with or without DPN compared to healthy con-
isfactory results. So far, there has been no trols, significantly higher in patients who have
reported pathologic study or nerve biopsy study DPN [58, 59] and even in the nerves that have
on DTRN.  However, we have biopsied thoracic normal results of electrophysiological studies
rootlets from individual cases and have seen peri- [59]. These evidences suggest structural abnor-
vascular inflammatory infiltrates (personal expe- malities, even subclinical, in diabetic nerves
rience). Furthermore, the similar clinical features increasing susceptibility to entrapment neuropa-
of abrupt onset of pain and weakness, associated thies. The causes of nerve enlargement are likely
weight loss, and monophasic course make it multifactorial and related to underlying meta-
likely that DTRN share a similar pathogenesis bolic derangement, oxidative stress and accumu-
with DCRPN and DLRPN and thereby should be lation of glycation products of sorbitol pathway
classified as a spectrum of DRPN similar to and inflammatory process described as an under-
DCRPN and DLRPN. lying mechanism in DPN [60]. The following are
the descriptions of each entrapment neuropathy
in association with diabetes mellitus. However, a
6 Entrapment Neuropathy detailed account of each entrapment neuropathy
is beyond the scope of this chapter and the read-
Entrapment neuropathies involving the median, ers are recommended to consult the other appro-
ulnar and fibular nerves, and the lateral cutaneous priate sources of references.
nerve of the thigh are commonly seen in diabetic
patients [54]. Diabetes mellitus has been recently
shown to be a risk factor of entrapment neuropa- 7 Median Mononeuropathy
thies in the upper extremity (median mononeu- Across the Wrist (MNW)
ropathy across the wrist (MNW) or carpal tunnel
syndrome (CTS) and ulnar mononeuropathy at MNW (also known as carpal tunnel syndrome) is
the elbow (UNE)) in a large longitudinal, the most common entrapment neuropathy in dia-
population-­based Swedish study [55]. The risk of betes mellitus [54]. Asymptomatic MNW with
developing entrapment neuropathy is higher in only abnormal electrophysiological findings has
diabetic patients who have diabetic sensorimotor been reported in 22% in type 1 diabetes mellitus
neuropathy (DPN), particularly in females and in and 29% in type 2 diabetes mellitus while symp-
type 2 diabetes mellitus [56]. An increased risk of tomatic MNW in 11% in type 1 diabetes mellitus
entrapment neuropathy in diabetes mellitus can and 6% in type 2 diabetes mellitus in a
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 173

community-­based longitudinal cohort of diabetic 454,987 patients undergoing carpal tunnel sur-
patients (the Rochester Diabetic Neuropathy gery [67]. Because underlying metabolic
Study) [1]. The prevalence of MNW in general derangement could make diabetic nerve suscep-
population has been estimated at 2–4% [61] but tible to entrapment at the pressure sites, attention
in diabetes mellitus, the prevalence has been to metabolic control is equally important in addi-
reported to be 14% in patients without DPN and tion to conventional management of MNW in
30% in those with DPN [62]. MNW occurs as a diabetic patients [54].
result of median nerve compression at the carpal
ligament [54]. MNW typically causes paresthesia
in the palmar surface of the thumb, second, third, 8 Ulnar Mononeuropathy
and the radial half of the fourth digits and, if Across the Elbow (UNE)
severe, weakness in the thumb abductor [63]. Not
uncommonly, paresthesia of the whole hand and UNE ranks second to MNW in prevalence in dia-
pain in the wrist are also described. These symp- betic patients with prevalence about 2.1% [54].
toms are precipitated by prolonged flexion or Similar to MNW, diabetes mellitus has been
extension of the wrist such as driving or during shown to be a risk factor for UNE [55]. The prev-
sleep [63]. The symptoms of MNW in diabetic alence of diabetes mellitus in UNE is about 6%
patients are similar to non-diabetic subjects but [68]. Most diabetic UNE cases are asymptomatic
coexisting symptoms of DPN (i.e., numbness/ or subclinical and are detected by electrophysio-
pain in the toes or feet) are usually present [64]. logical studies [69, 70]. Similar to MNW, UNE is
The diagnosis of MNW requires compatible elec- found more commonly in diabetic patients with
trodiagnostic abnormalities showing prolonged DPN [70]. The symptoms of UNE in diabetic
distal latencies in the median sensory or motor patients are similar to non-diabetic UNE.  Most
studies or slowing of the median conduction common symptoms are paresthesia or numbness
velocity across the wrist [63]. Recently the use of at the fifth digit, the medial half of the fourth digit
diagnostic high-resolution ultrasonographic stud- and the medial half of the volar surface of the
ies for MNW has become more widespread [65]. ipsilateral hand [71]. In a more severe case,
An ultrasound finding of an enlarged median intrinsic hand weakness and atrophy of the inter-
nerve in proximal carpal tunnel, edema around ossei resulting in clawing of the fourth and the
flexor tendons in cross-sectional images, and fifth digits and the Benediction posture or weak-
thickening of the flexor retinaculum could sug- ness of third palmar interosseus muscle resulting
gest CTS [65]. Management of symptomatic in Wartenburg’s sign can be seen [71]. In diabetic
MNW includes conservative treatment such as patients with severe UNE, many cases present
the use of wrist splint starting initially at night with motor weakness and atrophy without sen-
and escalating to the daytime and, if unsuccess- sory symptoms [72]. Inching studies showed that
ful, proceeding to local corticosteroid injection the most common location for diabetic UNE is at
and for more severe cases with weakness or atro- the retrocondylar groove [70]. The diagnosis of
phy of the thumb abductor, decompressive sur- UNE requires electrodiagnostic abnormalities
gery of the carpel tunnel [64]. A recent 5-year showing conduction block or slowing across the
follow-up study in diabetic patients who under- elbow [71]. However, in light of coexisting DPN
went carpal tunnel surgery showed significant in the majority of diabetic UNE patients, electro-
long-term neurophysiological and clinical physiologic abnormalities may not be clear cut
improvement in diabetic and non-diabetic due to slowing of the conduction velocity of the
patients alike and this improvement is indepen- ulnar nerve at the forearm segment. In this situa-
dent of the status of DPN in diabetic CTS patients tion electrophysiologic abnormalities may reveal
[66]. Diabetes mellitus was, however, found to be a non-localizing result and the diagnosis of UNE
an independent risk factor of infection following may require either inching study across the elbow
carpal tunnel surgery in a recent large cohort of [70] or a nerve ultrasound study [73] to make a
174 P. Thaisetthawatkul and P. J. B. Dyck

correct localization. A high-resolution ultrasound mellitus, FN can be seen as a result of diabetic


study of the ulnar nerve, similar to its utility in muscle infarct involving peroneal muscles [81].
MNW, has become an important diagnostic tool The diagnosis requires elevated creatine kinase and
in UNE [74]. Similar to an ultrasound study at the MRI of the lower extremity to demonstrate muscle
median nerve, a high-resolution ultrasound study infarct [81]. FN causes weakness of ipsilateral foot
at the ulnar nerve showed significant enlargement dorsiflexion and eversion but plantar dorsiflexion
in the cross-sectional area of the nerve in diabetic and inversion are spared (as these functions are
patients, particularly in patients with DPN [75]. controlled by the tibial nerve). Ipsilateral footdrop
This could also explain why UNE is also seen is a very common clinical presentation. The diag-
commonly in diabetic patients, particularly in nosis requires electrodiagnostic abnormalities
DPN patients. Management of symptomatic showing slowing or conduction block of the fibular
UNE in diabetic patients is similar to non-­diabetic motor study with or without axonal loss [78]. The
UNE. In a mild case, non-operative management major differential diagnosis for FN is L5 radicu-
in the form of wearing a soft elbow pad or avoid- lopathy, lumbosacral plexopathy (DLRPN) or sci-
ing prolonged elbow flexion may be considered atic mononeuropathy [82]. As some cases of FN
and for more severe cases and in the cases that can occur in patients with DPN, electrodiagnostic
fail conservative treatment, surgical intervention abnormalities for FN may not be definite as DPN is
may be considered [76]. Another form of ulnar a length-dependent axonal sensorimotor neuropa-
mononeuropathy seen in diabetic patients is ulnar thy and loss of either fibular motor response record-
neuropathy in the forearm [77]. Ulnar neuropathy ing at extensor digitorum brevis muscle or
in the forearm has similar clinical symptoms to superficial fibular sensory response or both indicat-
UNE but electrodiagnostic studies localize the ing axon loss is common in DPN. In these situa-
lesion in the forearm, i.e. conduction block or tions, fibular motor study recording at tibialis
abnormal temporal dispersion are seen between anterior muscle, which is a more proximal muscle,
the wrist and the elbow [77]. It is unclear if this if recordable, or a detailed needle electromyo-
type of ulnar neuropathy is part of DPN or it has graphic study may provide helpful diagnostic find-
a structural cause for it. ings [79]. A neuromuscular ultrasound is also
helpful in diagnosing FN demonstrating enlarge-
ment of nerve cross section area at the fibular head
9 Fibular Mononeuropathy [83]. Management of FN depends on the etiology
Across the Fibular Head (FN) but conservative treatment with the use of an ankle
brace and avoiding compression is the mainstay
FN is caused by compression of the fibular (pero- strategy. Surgical management is controversial and
neal) nerve at the bony prominence of the fibular should be limited to FN that has a clear structural
neck and is the most common entrapment neuropa- cause or severe FN that is prolonged and does not
thy of the lower limb [54]. FN commonly occurs recover with conservative approach [82].
after habitual leg crossing, prolonged hospitaliza-
tion, prolonged bedrest or during a surgical proce-
dure requiring general anesthesia [78]. FN often 10 Lateral Femoral Cutaneous
occurs when there is a large amount of weight loss Mononeuropathy [Meralgia
due to loss of subcutaneous fat padding around the Paresthetica (MP)]
common fibular nerve, such as severe malnutrition
or after having cancers [78]. Diabetes mellitus has MP is an entrapment syndrome that causes
been described as a precipitating factor in a large chronic neuropathic pain in the anterior and lat-
case series of FN, seen in 13 out of 103 FN cases eral thigh. It is the second most common entrap-
[79]. The prevalence of FN in diabetes mellitus is ment neuropathy in the lower extremity [84]. It is
unknown but FN was found in 13 out of 29 mono- caused by entrapment of the lateral femoral cuta-
neuropathies in diabetic patients [80]. In diabetes neous nerve of the thigh near the anterior supe-
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 175

rior iliac spine while passing beneath the inguinal treatment [78]. The symptoms in pregnant
ligament [78]. The symptoms of MP consist of patients usually resolve after delivery. Surgical
persistent or intermittent positive (tingling, decompression of the lateral femoral cutaneous
numbness, burning, stabbing or aching) or nega- nerve of the thigh is reserved for persistent pain
tive (sensory loss or hypoesthesia) symptoms after conservative management [78]. In very
without muscle weakness in unilateral or ­bilateral severe retractable cases neurectomy of lateral
anterolateral thighs [84]. The association between femoral cutaneous nerve has been performed and
MP and diabetes mellitus was shown in a popula- resulted in relief of pain [87].
tion-based study in Olmsted County Minnesota
[81]. In this study, the incidence of MP was esti-
mated to be seven times increased in diabetic 11 Cranial Neuropathies
patients compared to general population and that
MP patients are two times more likely to go on to Cranial mononeuropathies are uncommon mono-
develop diabetes mellitus in the future [85]. In neuropathies that care occasionally seen in dia-
addition, multivariate analysis showed that dia- betic patients, the frequency of which ranging
betes mellitus is an independent risk factor for from none in a population-based cohort of
MP [85]. Moreover, MP has been shown to be Rochester Diabetic Neuropathy Study [1] to
associated with obesity [85] and pregnancy [86]. 0.75% in a more recent hospital-based cohort
A femoral neuropathy or upper lumbar radicu- [89]. The most common diabetic cranial mono-
lopathy or upper lumbar plexopathy (DLRPN) neuropathy is oculomotor (CNIII) mononeuropa-
may present with similar symptoms [78]. The thy (0.35%) while facial (CN VII)
pathophysiology is likely to be compressive in mononeuropathy ranks second (0.21%) followed
most cases but in some retracted cases where by abducens (CN VI) mononeuropathy (0.15%)
neurectomy was performed and hence pathology and multiple cranial nerve palsies can be seen in
was available, inflammatory infiltrates, perineu- about 0.04% [89]. Trochlear (CN IV) mononeu-
rial thickening, hemosiderin-laden macrophages, ropathy has been only very rarely described in
and multifocal fiber loss were found raising the diabetic patients [90]. Diabetic patients who
possibility of inflammatory mechanisms in some develop cranial mononeuropathies usually have
[87]. The presence of muscle weakness or loss of older ages, prolonged duration of diabetes melli-
knee reflex makes MP unlikely but to exclude tus, type 2, poor control of blood glucose and co-­
those possibilities, detailed NCS/EMG in the morbidities [89].
lower extremities are required [78].
Electrodiagnostic abnormalities in MP may be
aided by performing the sensory conduction 12 Ocular Motor
study of the lateral femoral cutaneous nerve, Mononeuropathies (OMM)
which is technically difficult to do especially in
an obese subject because of anatomical variation. Despite its rarity, diabetic ophthalmoplegia
There are several methods of sensory studies of causes significant symptoms and anxiety to
the lateral femoral cutaneous nerve using either affected patients as it usually occurs abruptly
needle or surface stimulators or recorders [88]. causing diplopia and impairs daily activities
The abnormalities consistent with MP consist of significantly. Most commonly it involves CN
unilateral loss or decrease in the amplitude of the III or CN VI in isolation or less commonly a
lateral femoral cutaneous sensory study on the combination of these [91] but very rarely CN
ipsilateral side of the symptoms [88]. IV is involved. In a cohort of unselected cases
Management of MP consists of mainly conserva- of OMM, the frequency of diabetes mellitus
tive approach with losing weight and the use of was 7% [92] whereas in a large cohort of iso-
medications to relive neuropathic pain such as lated CN III palsy, diabetes mellitus was the
antidepressants, anticonvulsants, and local pain cause in 11% [93]. The onset of diabetic OMM
176 P. Thaisetthawatkul and P. J. B. Dyck

usually occurs in hours accompanied by ipsilat- cantly impaired pupillary reaction or aberrant
eral pain or headache, often refractory to anal- regeneration when there is CN III involvement.
gesics and diplopia [94]. The headache is either The underlying pathology causing OMM were
periorbital, retro-­orbital, frontal, temporal or elucidated by three clinic-­pathological studies
involves the entire side of the hemi-facial area [97–99], each of which were performed on a
[94]. Pain or headache can be seen in about diabetic patient who developed acute CN III
60% of cases and is most common in CN III palsy before death, showing either preservation
palsy [95]. It can occur about 1 week before the of axons [98] or axonal degeneration [97] lim-
onset of diplopia in about 35% of cases whereas ited to the central portion of the nerve trunk
65% have pain at the onset of diplopia [95]. The while the peripheral part of the nerve was
duration of pain can vary from a few days in a spared. In addition, there were extensive patho-
mild case to more than a month in a severe case logical changes seen in the intraneural arteri-
[95]. The other symptom includes ptosis (in CN oles or small arteries consisting of hyalinization
III palsy) but severe pupillary dysfunction is of the vessel walls and obliteration or marked
uncommon. In diabetic CN III palsy, impaired narrowing of the lumen without inflammation
pupillary reaction can be seen in up to 50% and [97–99]. The large arteries supplying CN III,
in about 60% of these, the impairment can be on the contrary, were found to be relatively
bilateral suggesting an accompanying diabetic unremarkable [97–99]. The pathological
autonomic neuropathy [93]. Even though changes were seen only in a very short segment
impaired pupillary reaction can be seen in dia- of CN III, two cases in the intracavernous por-
betic CN III palsy, anisocoria greater than tion, presumptively a watershed area of blood
2 mm is very rare [93]. Similarly, a fixed pupil supply [97, 98]. The underlying mechanism of
ipsilateral to the symptom with opposite normal diabetic CN III palsy was postulated to be isch-
pupil is very rare in diabetic CN III palsy while emia from microvascular origin [97–99].
this phenomenon can be seen in 61% of CN III Pupillary sparing is explained by the peripheral
palsy from a posterior communicating artery localization of the autonomic fibers supplying
aneurysm [93]. Diabetes mellitus is among the the pupils thereby escaping the ischemic injury
most common cause of pupil sparing CN III [100]. Microvascular ischemia as the cause of
palsy [93]. The implication of this matter lies in diabetic OMM is also supported by findings of
the necessity to differentiate diabetic CN III significant association between ischemic OMM
palsy from the other compressive structural and the risk factors for cardiovascular diseases
causes such as a posterior communicating in two studies [96, 101]. Moreover, diabetic
artery aneurysm or a tumor such as a pituitary OMM has been found to be an independent risk
tumor. In a large cohort of patients with iso- factor of ischemic stroke in type 2 diabetes
lated CN III palsy, diabetes mellitus was found mellitus in one population-based study [102].
to be the cause of CN III palsy in only 11%— However, the occurrence of diabetic retinopa-
another 6% of patients were diabetic and had thy was found to be significantly less common
another cause of CN III palsy including isch- in patients who had diabetic OMM than who
emic stroke such as midbrain infarct, infection had not suggesting a different pathophysiologic
or a posterior communicating artery aneurysm mechanism for these two microvascular com-
[93]. In another large prospective cohort of plications [90]. The course and prognosis of
patients with presumed benign CN III, IV, and diabetic OMM is self-recovery, either complete
VI palsies, a structural cause was identified in or partial in most cases [94]. In a large,
17% [96]. These findings underscore the need population-­based study of acquired CN III
of neuroimaging studies to identify other pos- palsy, 95% of microvascular CN III palsy had
sible structural lesions causing OMM in dia- complete while the rest had incomplete, yet
betic patients, particularly when there are signs substantial, recovery [103]. On the contrary,
suggesting a structural lesion such as signifi- CN III palsy from the other causes such as post-
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 177

surgery, trauma, aneurysm or malignancy has tizing external otitis [112], and mucormycosis in
only about 20–35% chance of complete recov- the parotid gland [113, 114]. These patients usu-
ery [103]. The time to recovery is usually within ally have additional symptoms and signs sugges-
4–6 weeks [94]. Management of diabetic OMM tive of local infections such as local facial
is conservative management and control of car- swelling, otalgia, otorrhea, local tenderness,
diovascular risk factors such as pain manage- fever, and crepitation over the skin [110–114].
ment, blood glucose and lipid control, Facial palsy has been described as part of multi-
management of blood pressure, smoking cessa- ple cranial neuropathies (polyneuritis cranialis)
tion, weight reduction, and the use of eye pad to in diabetic patients [115]. The treatment for
reduce diplopia or adhesive tape for ptosis [94]. Bell’s palsy is oral corticosteroid, usually
60–80  mg/day given in a short course over a
week, preferably started within 72  h after the
13 Facial Paralysis onset [104, 109]. A combination of corticosteroid
and anti-viral agent may be considered in a severe
Facial weakness is another common cranial neu- case but the benefit has been found to be marginal
ropathy seen in diabetic patients [89]. Idiopathic in clinical trials but the risk is minimal [104,
CN VII palsy or Bell’s palsy is the most common 109]. The use of anti-viral agent alone is not rec-
cause of facial weakness, accounting for up to ommended due to lack of benefit over the pla-
75% of cases [104]. Diabetes mellitus is more cebo [104, 109]. The course and prognosis of
prevalent in Bell’s palsy than in general popula- Bells’ palsy is that of self-improvement with
tion, ranging from 10% to 30% among many about 70% having complete recovery and about
studies, and is thought to be an important co-­ 15% having moderate to severe residual deficit
morbidity in the type of facial paralysis [105– [104]. The use of corticosteroid may help the
108] even though routine screening for diabetes most severely affected 30% group (who do not
mellitus is not yet recommended when a patient have a full recovery) to achieve a full recovery in
with Bells’ palsy is encountered at an initial visit about one-third to one-half [104]. The presence
[109]. Bells’ palsy is associated with swelling of diabetes mellitus does not seem to affect the
and compression of the facial nerve at the level of recovery of Bell’s palsy [116]. The severity of
the geniculate ganglion within the meatal seg- Bell’s palsy at the time of maximum deficit is an
ment of the nerve [104]. The cause of Bell’s palsy adverse prognostic factor [94]. Electrophysiologic
is likely from non-specific inflammation, with assessment of the facial motor study can predict
possible contribution in some cases from herpes poor prognosis in that if the facial compound
viral infection [104]. The symptoms consist of muscle action potential (CMAP) is less than 10%
unilateral facial paralysis, facial and retro-­ of the lower limit of normal, the chance of having
auricular pain, altered taste, intolerance to loud meaningful recovery is very unlikely and if the
noise (hyperacusis), and sensory symptoms in facial CMAP is more than 30% of lower normal
some cases [104]. Dry eyes, dry mouth, and croc- limit, the chance of having good recovery is
odile tear syndrome (lacrimation from salivary excellent [94]. In case of severe facial weakness
stimuli) can also be seen [104]. The other causes with guarded chance of recovery, the role of sur-
of facial paralysis that needs to be differentiated gical decompression is still controversial [104,
from typical Bell’s palsy include trauma, 109].
varicella-­
zoster infection (Ramsay Hunt syn-
drome), neoplasm, Lyme disease, and sarcoidosis Disclosure Statement  Pariwat Thaisetthawatkul MD has
[94]. In diabetic patients in particular, certain nothing to disclose.
malignant infections may present predominantly P. James B. Dyck MD has nothing to disclose.
with facial paralysis and needs to be excluded
such as candidal abscess of the parotid gland
[110], cervical necrotizing fasciitis [111], necro-
178 P. Thaisetthawatkul and P. J. B. Dyck

References 20. Dyck PJB, Windebank AJ.  Diabetic and nondia-


betic lumbosacral radiculoplexus neuropathies: new
insights into pathophysiology and treatment. Muscle
1. Dyck PJ, Kratz KM, Karnes JL, et  al. The preva- Nerve. 2002;25:477–91.
lence by staged severity of various types of dia- 21. Dyck PJB, Norell JE, Dyck PJ.  Non-diabetic lum-
betic neuropathy, retinopathy, and nephropathy in bosacral radiculoplexus neuropathy: natural history,
a population-based cohort: the Rochester Diabetic outcome and comparison with the diabetic variety.
Neuropathy Study. Neurology. 1993;43:817–24. Brain. 2001;124:1197–207.
2. Tracy JA, Dyck PJB.  The spectrum of diabetic 22. Ng PS, Dyck PJB, Laughlin RS, et al. Lumbosacral
neuropathies. Phys Med Rehabil Clin N Am. radiculoplexus neuropathy: Incidence and the
2008;19:1–26. association with diabetes mellitus. Neurology.
3. Thaisetthawatkul P, Dyck PJB.  Cervical and lum- 2019;92:e1188–94. https://doi.org/10.1212/
bosacral radiculoplexus neuropathies. In: Rajabally WNL.0000000000007020. Epub 2019 Feb 13.
Y, editor. Dysimmune neuropathies. San Diego: 23. Dyck PJB, Thaisetthawatkul P. Lumbosacral plexop-
Elsevier/Academic Press; 2020. p. 199–224. athy. Continuum (Minneap Minn). 2014;20:1343–58.
4. Sinnreich M, Taylor BV, Dyck PJB.  Diabetic neu- 24. Garces-Sanchez M, Laughlin RS, Dyck PJB, et  al.
ropathies. Classification, clinical features, and patho- Painless diabetic motor neuropathy: a variant of dia-
physiological basis. Neurologist. 2005;11:63–79. betic lumbosacral radiculoplexus Neuropathy? Ann
5. Dyck PJB, Norell JE, Dyck PJ. Microvasculitis and Neurol. 2011;69:1043–54.
ischemia in diabetic lumbosacral radiculoplexus 25. Pinto MV, Ng PS, Howe BM, Laughlin RS, Thapa
neuropathy. Neurology. 1999;53:2113–21. P, Dyck PJ, Dyck PJB. Lumbosacral radiculo-
6. Massie R, Mauermann ML, Staff NP, et al. Diabetic plexus neuropathy: neurologic outcomes and sur-
cervical radiculoplexus neuropathy: a distinct syn- vival in a population-based study. Neurology.
drome expanding the spectrum of diabetic radiculo- 2021;96(16):e2098–e2108. https://doi.org/10.1212/
plexus neuropathies. Brain. 2012;135:3074–88. WNL.0000000000011799.
7. Garland H, Taverner D.  Diabetic myelopathy. Br 26. Llewelyn JG, Thomas PK, King RH.  Epineurial
Med J. 1953;1:1405–8. microvasculitis in proximal diabetic neuropathy. J
8. Garland H.  Diabetic amyotrophy. Br Med J. Neurol. 1998;245:159–65.
1955;2:1287–90. 27. Dyck PJB, Engelstad J, Norell J, Dyck
9. Skanse B, Gydell K. A rare type of femoral-sciatic PJ.  Microvasculitis in non-diabetic lumbosacral
neuropathy in diabetes mellitus. Acta Med Scand. radiculoplexus neuropathy (LSRPN): similarity to
1956;155:463–8. the diabetic variety (DLSRPN). J Neuropathol Exp
10. Calverly JR, Mulder DW.  Femoral neuropathy. Neurol. 2000;59:525–38.
Neurology. 1960;10:963–7. 28. Kawamura N, Dyck PJB, Schmeichel AM, et  al.
11. Raff MC, Sangalang V, Asbury AK. Ischemic mono- Inflammatory mediators in diabetic and non-­diabetic
neuropathy multiplex in association with diabetes lumbosacral radiculoplexus neuropathy. Acta
mellitus. Neurology. 1968;18:284. Neuropathol. 2008;115:231–9.
12. Williams IR, Mayer RF. Subacute proximal diabetic 29. Holtzman DM, Davis RE, Greco CM. Lumbosacral
neuropathy. Neurology. 1976;26:108–16. plexopathy secondary to perirectal abscess in a patient
13. Asbury AK.  Proximal diabetic neuropathy. Ann with HIV infection. Neurology. 1989;39:1400–1.
Neurol. 1977;2:179–80. 30. Schaller MA, Wicke F, Foerch C, et  al. Central
14. Chokroverty S, Reyes MG, Rubino FA.  Bruns-­ nervous system tuberculosis: etiology, clinical
Garland syndrome of diabetic amyotrophy. Trans manifestations and neuroradiological features. Clin
Am Neurol Assoc. 1977;102:173–7. Neuroradiol. 2019;29:3–18.
15. Barohn RJ, Sahenk Z, Warmolts JR, et al. The Bruns-­ 31. Chahin N, Temesgen Z, Kurtin PJ, et al. HIV lum-
Garland syndrome (diabetic amyotrophy). Revisited bosacral radiculoplexus neuropathy mimicking
100 years later. Arch Neurol. 1991;48:1130–5. lymphoma: diffuse infiltrative lymphocytosis syn-
16. Bastron JA, Thomas JE.  Diabetic polyradiculopa- drome (DILS) restricted to nerve? Muscle Nerve.
thy: clinical and electromyographic findings in 105 2010;41:276–82.
patients. Mayo Clin Proc. 1981;56:725–32. 32. Staff NP, Engelstad J, Klein CJ, et al. Post-surgical
17. Bradley WG, Chad D, Verghese JP, et  al. Painful inflammatory neuropathy. Brain. 2010;133:2866–80.
lumbosacral plexopathy with elevated erythrocyte 33. Dyck PJ, O’Brien P, Bosch EP, et  al. The multi-­
sedimentation rate: a treatable inflammatory syn- centre double-blind controlled trial of IV
drome. Ann Neurol. 1984;15:457–64. methylprednisolone in diabetic lumbosacral radicu-
18. Krendel DA, Costigan DA, Hopkins LC. Successful loplexus neuropathy. Neurology. 2006;66(5 Suppl
treatment of neuropathies in patients with diabetes 2):A191.
mellitus. Arch Neurol. 1995;52:1053–61. 34. Dyck PJB, Norell JE, Dyck PJ. Methylprednisolone
19. Said G, Lacroix C, Lozeron P, et  al. Inflammatory may improve lumbosacral radiculoplexus neuropa-
vasculopathy in multifocal diabetic neuropathy. thy. Can J Neurol Sci. 2001;28:224–7.
Brain. 2003;126:376–85.
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 179

35. Kilfoyle D, Kelkar P, Parry GJ.  Pulsed methyl- 54. Massey EW.  Diabetic truncal mononeuropathy:
prednisolone is a safe and effective treatment for electromyographic evaluation. Acta Diabetol Lat.
diabetic amyotrophy. J Clin Neuromuscul Dis. 1980;17:269–72.
2003;4:168–70. 55. Vinik A, Mehrabyan A, Colen L, et al. Focal entrap-
36. Thaisetthawatkul P, Dyck PJB. Treatment of diabetic ment neuropathies in diabetes. Diabetes Care.
and nondiabetic lumbosacral radiculoplexus neurop- 2004;27:1783–8.
athy. Curr Treat Options Neurol. 2010;12:95–9. 56. Rydberg M, Zimmerman M, Gottsäter A, et  al.
37. Chan YC, Lo YL, Chan ES.  Immunotherapy for Diabetes mellitus as a risk factor for compres-
diabetic amyotrophy. Cochrane Database Syst Rev. sion neuropathy: a longitudinal cohort study from
2012;6:CD006521. southern Sweden. BMJ Open Diabetes Res Care.
38. Auche MB. Des alterations des nerfs peripheriques. 2020;8:e001298.
Arch Med Exp Anat pathol. 1890;2:635. 57. Moon HI, Shin J, Kim YW, et al. Diabetic polyneu-
39. Parsonage MJ, Turner JW.  Neuralgic amyotrophy; ropathy and the risk of developing carpal tunnel
the shoulder-girdle syndrome. Lancet. 1948;1:973–8. syndrome: a nationwide, population-based study.
40. Tsairis P, Dyck PJ, Mulder DW. Natural history of Muscle Nerve. 2020;62:208–13.
brachial plexus neuropathy. Report on 99 patients. 58. Mojaddidi MA, Ahmed MS, Ali R, et al. Molecular
Arch Neurol. 1972;27:109–17. and pathological studies in the posterior interosseous
41. van Alfen N, van Engelen BG.  The clinical spec- nerve of diabetic and non-diabetic patients with car-
trum of neuralgic amyotrophy in 246 cases. Brain. pal tunnel syndrome. Diabetologia. 2014;57:1711–9.
2006;129:438–50. 59. Breiner A, Qrimli M, Ebadi H, et  al. Peripheral
42. Kikta DG, Breuer AC, Wilbourn AJ.  Thoracic root nerve high-resolution ultrasound in diabetes. Muscle
pain in diabetes: the spectrum of clinical and elec- Nerve. 2017;55:171–8.
tromyographic findings. Ann Neurol. 1982;11:80–5. 60. Pitarokoili K, Kerasnoudis A, Behrendt V, et  al.
43. Ellenberg M.  Diabetic truncal mononeuropathy—a Facing the diagnostic challenge: nerve ultrasound
new clinical syndrome. Diabetes Care. 1978;1:10–3. in diabetic patients with neuropathic symptoms.
44. Longstreth GF, Newcomer AD.  Abdominal pain Muscle Nerve. 2016;54:18–24.
caused by diabetic radiculopathy. Ann Intern Med. 61. Figueroa-Romero C, Sadidi M, Feldman
1977;86:166–8. EL.  Mechanisms of disease: the oxidative stress
45. Chaudhuri KR, Wren DR, Werring D, et  al. theory of diabetic neuropathy. Rev Endocr Metab
Unilateral abdominal muscle herniation with pain: a Disord. 2008;9:301–14.
distinctive variant of diabetic radiculopathy. Diabet 62. Mediouni Z, Bodin J, Dale AM, et  al. Carpal tun-
Med. 1997;14:803–7. nel syndrome and computer exposure at work in
46. O’Connor RC, Andary MT, Russo RB, et  al. two large complementary cohorts. BMJ Open.
Thoracic radiculopathy. Phys Med Rehabil Clin N 2015;5:e008156.
Am. 2002;13:623–44. 63. Perkins BA, Olaleye D, Bril V.  Carpal tunnel syn-
47. Alshekhlee A, Tay E, Buczek M, Shakir ZA, et al. drome in patients with diabetic polyneuropathy.
Herpes zoster with motor involvement: discordance Diabetes Care. 2002;25:565–9.
between the distribution of skin rash and localiza- 64. Preston DC, Shapiro BE. Median neuropathy at the
tion of peripheral nervous system dysfunction. J Clin wrist. In: Electromyography and neuromuscular
Neuromuscul Dis. 2011;12:153–7. disorders: clinical-electrophysiologic-ultrasound
48. Mormont E, Esselinckx W, De Ronde T, et  al. correlations. 4th ed. Philadelphia: Elsevier; 2021.
Abdominal wall weakness and lumboabdominal p. 323–57.
pain revealing neuroborreliosis: a report of three 65. Smith BE.  Focal and entrapment neuropathies.
cases. Clin Rheumatol. 2001;20:447–50. Handb Clin Neurol. 2014;126:31–43.
49. Hamodat H, Tran A.  Neurosarcoidosis resulting in 66. Yoshii Y, Zhao C, Amadio PC. Recent advances in
thoracic radiculopathy: a case report. J Med Case ultrasound diagnosis of carpal tunnel syndrome.
Rep. 2019;13:130. Diagnostics (Basel). 2020;10:596.
50. Perrini P, Benedetto N, Buccoliero AM, et al. Thoracic 67. Thomsen NOB, Andersson GS, Björk J, et  al.
radiculopathy from a paravertebral mesothelial cyst. Neurophysiological recovery 5 years after carpal
Acta Neurochir (Wien). 2006;148:989–91. tunnel release in patients with diabetes. Muscle
51. Kim Y, Kim S, Oh K, et al. Thoracic radiculopathy Nerve. 2017;56:E59–64.
as initial symptoms of hepatocellular carcinoma: a 68. Werner BC, Teran VA, Deal DN.  Patient-related
case report. Medicine (Baltimore). 2018;97:e11635. risk factors for infection following open carpal tun-
52. Choi HE, Shin MH, Jo GY, et  al. Thoracic radicu- nel release: an analysis of over 450,000 Medicare
lopathy due to rare causes. Ann Rehabil Med. patients. J Hand Surg Am. 2018;43:214–9.
2016;40:534–9. 69. Mondelli M, Aretini A, Rossi S.  Ulnar neuropathy
53. Fitzpatrick MO, Goyal K, Johnston RA.  Thoracic at the elbow in diabetes. Am J Phys Med Rehabil.
radiculopathy caused by a myodil cyst. Br J 2009;88:278–85.
Neurosurg. 2000;14:351–3. 70. Rota E, Zavaroni D, Parietti L, et al. Ulnar entrap-
ment neuropathy in patients with type 2 diabetes
180 P. Thaisetthawatkul and P. J. B. Dyck

mellitus: an electrodiagnostic study. Diabetes Res 87. van Slobbe AM, Bohnen AM, Bernsen RM, et  al.
Clin Pract. 2014;104:73–8. Incidence rates and determinants in meralgia pares-
71. Jang JE, Kim YT, Park BK, et al. Subclinical ulnar thetica in general practice. J Neurol. 2004;251:294–7.
neuropathy at the elbow in diabetic patients. Ann 88. Berini SE, Spinner RJ, Jentoft ME, et  al. Chronic
Rehabil Med. 2014;38:64–71. meralgia paresthetica and neurectomy: a clinical
72. Preston DC, Shapiro BE.  Ulnar neuropathy at the pathologic study. Neurology. 2014;82:1551–5.
elbow. In: Electromyography and neuromuscular 89. Oh S.  Clinical electromyography. 3rd ed.
disorders: clinical-electrophysiologic-ultrasound Philadelphia: Lippincott Williams and Wilkins; 2003.
correlations. 4th ed. Philadelphia: Elsevier; 2021. 90. Greco D, Gambina F, Pisciotta M, et  al. Clinical
p. 372–401. characteristics and associated comorbidities in dia-
73. Schady W, Abuaisha B, Boulton AJ.  Observations betic patients with cranial nerve palsies. J Endocrinol
on severe ulnar neuropathy in diabetes. J Diabetes Invest. 2012;35:146–9.
Complications. 1998;12:128–32. 91. Trigler L, Siatkowski RM, Oster AS, et  al.
74. Pelosi L, Yong V, Blumhardt LD.  Utility of nerve Retinopathy in patients with diabetic ophthalmople-
ultrasound in ulnar neuropathy with abnormal non-­ gia. Ophthalmology. 2003;110:1545–50.
localizing electrophysiology in diabetes mellitus. 92. Greco D, Gambina F, Maggio F.  Ophthalmoplegia
Neurophysiol Clin. 2020;50:387–90. in diabetes mellitus: a retrospective study. Acta
75. Schertz M, Mutschler C, Masmejean E, et  al. Diabetol. 2009;46:23–6.
High-resolution ultrasound in etiological evalua- 93. Rush JA, Younge BR. Paralysis of cranial nerves III,
tion of ulnar neuropathy at the elbow. Eur J Radiol. IV, and VI. Cause and prognosis in 1,000 cases. Arch
2017;95:111–7. Ophthalmol. 1981;99:76–9.
76. Chen J, Wang CL, Wu S, et  al. The feasibility of 94. Keane JR.  Third nerve palsy: analysis of 1400
using high-resolution ultrasonography to assess personally-examined inpatients. Can J Neurol Sci.
ulnar nerve in patients with diabetes mellitus. J 2010;37:662–70.
Ultrason. 2017;17:160–6. 95. Smith BE. Cranial neuropathy in diabetes mellitus.
77. Nakashian MN, Ireland D, Kane PM. Cubital tunnel In: Dyck PJ, Thomas PK, editors. Diabetic neu-
syndrome: current concepts. Curr Rev Musculoskelet ropathy. 2nd ed. Philadelphia: WB Saunders; 1999.
Med. 2020;13:520–4. p. 457–67.
78. Acosta JA, Hoffman SN, Raynor EM, et  al. Ulnar 96. Wilker SC, Rucker JC, Newman NJ, et  al. Pain in
neuropathy in the forearm: a possible complication ischaemic ocular motor cranial nerve palsies. Br J
of diabetes mellitus. Muscle Nerve. 2003;28:40–5. Ophthalmol. 2009;93:1657–9.
79. Katirji MB.  Compressive and entrapment neu- 97. Tamhankar MA, Biousse V, Ying GS, et al. Isolated
ropathies of the lower extremity. In: Katirji MB, third, fourth, and sixth cranial nerve palsies from
Kaminski HJ, Ruff RL, editors. Neuromuscular presumed microvascular versus other causes: a pro-
disorders in clinical practice. 2nd ed. New  York: spective study. Ophthalmology. 2013;120:2264–9.
Springer; 2014. p. 903–52. 98. Dreyfus PM, Hakim S, Adams RD.  Diabetic oph-
80. Katirji MB, Wilbourn AJ. Common peroneal mono- thalmoplegia; report of case, with postmortem
neuropathy: a clinical and electrophysiologic study study and comments on vascular supply of human
of 116 lesions. Neurology. 1988;38:1723–8. oculomotor nerve. AMA Arch Neurol Psychiatry.
81. Mulder DW, Lambert EH, Bastron JA, et al. The neu- 1957;77:337–49.
ropathies associated with diabetes mellitus. A clini- 99. Asbury AK, Aldredge H, Hershberg R, et  al.
cal and electromyographic study of 103 unselected Oculomotor palsy in diabetes mellitus: a clinico-­
diabetic patients. Neurology. 1961;11:275–84. pathological study. Brain. 1970;93:555–66.
82. Desmottes MC, Brehier Q, Bertolini E, et  al. 100. Weber RB, Daroff RB, Mackey EA.  Pathology of
Compression of the common peroneal nerve due to oculomotor nerve palsy in diabetics. Neurology.
peroneal muscle infarction in a patient with diabetes. 1970;20:835–8.
Joint Bone Spine. 2019;86:265–6. 101. Sunderland S, Hughes ES.  The pupillo-constrictor
83. Bowley MP, Doughty CT.  Entrapment neuropa- pathway and the nerves to the ocular muscles in
thies of the lower extremity. Med Clin North Am. man. Brain. 1946;69:301–9.
2019;103:371–82. 102. Jung JS, Kim DH. Risk factors and prognosis of iso-
84. Hobson-Webb LD, Juel VC.  Common entrap- lated ischemic third, fourth, or sixth cranial nerve
ment neuropathies. Continuum (Minneap Minn). palsies in the Korean population. J Neuroophthalmol.
2017; 23(2, Selected Topics in Outpatient 2015;35:37–40.
Neurology):487–511. 103. Park SJ, Yang HK, Byun SJ, et al. Risk of ischemic
85. Seror P, Seror R. Meralgia paresthetica: clinical and stroke after third, fourth, and sixth cranial nerve pal-
electrophysiological diagnosis in 120 cases. Muscle sies in type 2 diabetes. J Diabetes. 2019;11:379–85.
Nerve. 2006;33:650–4. 104. Fang C, Leavitt JA, Hodge DO, et  al. Incidence
86. Parisi TJ, Mandrekar J, Dyck PJB, et  al. Meralgia and etiologies of acquired third nerve palsy using
paresthetica: relation to obesity, advanced age, and a population-based method. JAMA Ophthalmol.
diabetes mellitus. Neurology. 2011;77:1538–42. 2017;135:23–8.
Asymmetric Diabetic Neuropathy: Radiculoplexus Neuropathies, Mononeuropathies, and Cranial… 181

105. Glass GE, Tzafetta K.  Bell’s palsy: a summary of 112. Al-Ammar A, Maqbool MS.  Cervical necrotizing
current evidence and referral algorithm. Fam Pract. fasciitis with facial nerve paralysis. J Laryngol Otol.
2014;31:631–42. 2004;118:573–5.
106. Adour KK, Bell DN. Incidence of diabetes mellitus 113. Franco-Vidal V, Blanchet H, Bebear C, et  al.
in Bell’s palsy. Lancet. 1971;2:168. Necrotizing external otitis: a report of 46 cases. Otol
107. Adour K, Wingerd J, Doty HE. Prevalence of con- Neurotol. 2007;28:771–3.
current diabetes mellitus and idiopathic facial paral- 114. Chandu A, MacIsaac RJ, MacGregor DP, Campbell
ysis (Bell’s palsy). Diabetes. 1975;24:449–51. MC, Wilson MJ, Bach LA.  A case of mucormy-
108. Brandenburg NA, Annegers JF.  Incidence and risk cosis limited to the parotid gland. Head Neck.
factors for Bell’s palsy in Laredo, Texas: 1974-1982. 2005;27:1108–11.
Neuroepidemiology. 1993;12:313–25. 115. Gochhait D, Alexender A, Dehuri P, et al. Mucormycotic
109. Zhao H, Zhang X, Tang YD, Zhu J, Wang XH, Li parotitis and otitis causing facial nerve palsy in a dia-
ST.  Bell’s palsy: clinical analysis of 372 cases betic patient. Cytopathology. 2018;29:582–4.
and review of related literature. Eur Neurol. 116. Eshbaugh CG, Siatkowski RM, Smith JL, et  al.
2017;77:168–72. Simultaneous, multiple cranial neuropathies in dia-
110. Albers JR, Tamang S. Common questions about Bell betes mellitus. J Neuroophthalmol. 1995;15:219–24.
palsy. Am Fam Physician. 2014;89:209–12. 117. Şevik Eliçora S, Erdem D. Does type 2 diabetes mel-
111. Marioni G, Rinaldi R, de Filippis C, et  al. litus affect the healing of Bell’s palsy in adults? Can
Candidal abscess of the parotid gland associated J Diabetes. 2018;42:433–6.
with facial nerve paralysis. Acta Otolaryngol.
2003;123:661–3.
Motor Neuropathy in Diabetes

Karolina Snopek Khan and Henning Andersen

1 Introduction imaging (MRI), and histological evaluation based


on muscle biopsies. However, in a clinical set-
Distal symmetric polyneuropathy (DSPN) is the ting, these methods are only very rarely imple-
most common clinical presentation of diabetic mented, and are mainly used for research
neuropathy [1]. Motor and sensory nerves are purposes. Identifying motor neuropathy in diabe-
concomitantly affected early in the course of tes is of importance as it may lead to motor dys-
DSPN when examined by nerve conduction stud- function [5] and has serious clinical consequences
ies (NCS) [2]. However, clinically motor neurop- with an increased risk of foot ulcers [4], unstable
athy first becomes apparent later in the course of gait [6], postural instability [7], frequent falls [8,
DSPN [3]. This more advanced stage of DSPN 9], and impaired joint mobility [10, 11] leading to
has been denoted type 2b by Dyck et al., defined increased morbidity. Currently, there are no spe-
by the inability to stand on heels [4]. In cific treatments for motor neuropathy. However,
population-­based studies of individuals with type recent evidence has emerged showing benefits
1 and type 2 diabetes, this level of severe DSPN from training interventions for motor dysfunction
was reported in only 6 and 1% of patients, respec- in individuals with DSPN [12–14]. In recent
tively [4]. In recent years, there has been growing years new evidence suggests the concomitant
interest in motor dysfunction and muscle weak- existence and development of a myopathy in dia-
ness in diabetes and in relation to DSPN. Motor betes [15]. It has been speculated that these
neuropathy leads to muscle weakness and atro- changes may further explain the observed motor
phy, which ultimately results in motor impair- dysfunction and muscle weakness seen in indi-
ment. Motor neuropathy can be diagnosed based viduals with diabetes.
on a combination of clinical tests and In this chapter, we deal with all aspects of
NCS.  Several methods have been applied to motor neuropathy, including clinical presenta-
assess muscle size and quality, including imaging tion, pathophysiology, muscle strength, diagnos-
techniques such as ultrasonography (USG), com- tic tools, functional consequences of motor
puted tomography (CT) and magnetic resonance impairment, and effects of exercise.

K. S. Khan · H. Andersen (*)


Department of Neurology, Aarhus University
Hospital, University of Aarhus, Aarhus, Denmark
e-mail: hena@clin.au.dk

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 183
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_11
184 K. S. Khan and H. Andersen

2 Clinical Presentation viduals with diabetes, including mononeuropa-


thies, radiculopathies, and immune-mediated
In DSPN, sensory signs usually precede clinical neuropathies (Table 1). DSPN is a diagnosis of
motor dysfunction. Individuals with DSPN and exclusion, and therefore screening for other
motor involvement usually do not complain of causes of polyneuropathy and a thorough medi-
muscle weakness until more advanced stages of cal history should be obtained. Motor neuropa-
the disease, and therefore it may go unnoticed for thies in diabetes with an asymmetrical pattern,
a long time. proximal distribution, motor predominant
When motor dysfunction becomes apparent, involvement or an acute onset, should be con-
signs of sensory abnormalities are usually evi- sidered atypical, requiring thorough examina-
dent at the clinical examination. As with sensory tions for other causes such as inherited
disturbances, motor symptoms and signs ini- polyneuropathies, motor neuron disease and
tially present distally in the lower extremities acquired immune-mediated polyneuropathies
and progress more proximally. Other less fre- (Table  1). Furthermore, age-related muscle
quent presentations of diabetic neuropathy weakness due to atrophy and sarcopenia are
should be kept in mind when examining indi- other etiologies that need consideration.

Table 1  Atypical diabetic neuropathies and other motor neuropathies


Distribution and
characteristics Clinical features Progression Electrodiagnostic studies
Diabetic
radiculoplexus
neuropathies:
 Lumbosacral Often unilateral Pain followed by Subacute Predominantly axonal
or asymmetric, proximal weakness of degeneration with segmental
may present the lower limb demyelination
with muscles often
symmetrical combined with weight
distribution loss
 Cervicobrachial Asymmetric May involve proximal Predominantly axonal
upper extremity degeneration with segmental
muscles demyelination
 Thoracic May be Pain and dysesthesias EMG of paraspinal muscles
radiculoneuropathy symmetric, involving the chest/ shows signs of denervation
involving abdominal wall can
multiple lead to respiratory
dermatomes weakness
Mononeuropathies Asymmetric Cranial nerve palsies Slow Demyelinating at typical
(oculomotor nerve sites of compression
and facial nerve), in
limbs most
commonly: median,
ulnar and peroneal
nerve
Multifocal motor Asymmetric, Distal muscle Slow Conduction block followed
neuropathy pure motor weakness by axonal degeneration
Motor neuron disease Initially Asymmetric limb Progresses at Initially normal ENG. As
asymmetric, weakness in 80%, and different atrophy becomes severe,
pure motor bulbar weakness in rates, usually axonal loss is seen. EMG
20%, and 1–3% with fast discloses signs of acute and
respiratory weakness chronic muscle denervation
in several muscles and body
regions
Motor Neuropathy in Diabetes 185

Table 1 (continued)
Distribution and
characteristics Clinical features Progression Electrodiagnostic studies
Inflammatory/ Symmetric, AIDP: Hypo- or Acute AIDP: demyelinating with
immune-mediated muscle areflexia sural sparring
Guillain Barre weakness AMAN: Distal and AMAN: motor axonal
Syndrome proximal muscle AMSAN: sensory and motor
weakness axonal
AMSAN: Distal and
proximal muscle
weakness
Chronic inflammatory Symmetric Distal and proximal Subacute and Demyelinating
demyelinating muscle weakness reoccurring
polyradiculoneuropathy
Radiculopathy Asymmetric Depending on Acute ENG: usually normal
location, characterized EMG: axonal denervation
by pain, muscle
weakness, numbness,
and tingling sensation
Hereditary:
  CMT type 1 Symmetric Distal to proximal Slow CMT1: Demyelinating
  CMT type 2 muscle weakness, CMT2: Axonal
severe distal muscle
atrophy, and high foot
arch

Complaints and clinical signs gradually 3 Pathophysiology


become more evident as the disease slowly pro-
gresses. When motor neuropathy becomes clini- The pathophysiology of motor neuropathy
cally apparent, complaints of symptoms from the remains unknown. The muscle weakness in
lower extremities are typical, such as weakness DSPN can be attributed to both neurogenic and
of the feet and legs, unstable gait, and difficulty myopathic abnormalities in the distal muscles of
walking. In the functional and clinical examina- the lower limbs, which may contribute to
tion, individuals usually present with an inability decreased muscle quality and abnormal muscle
to stand on heels, decreased strength at the toes structure [15].
and ankles, decreased or absent distal reflexes, In DSPN, there is a combination of axonal
pes cavus, muscle twitching, and muscle cramp- degeneration and demyelination affecting both
ing. In the advanced stages of DSPN, proximal motor and sensory nerves. Loss of motor axons
muscles in the lower extremities and the thighs leads to denervation of muscle fibers, causing
and even in the upper extremities may become muscle weakness, muscular atrophy [16], and
involved. increased fatigability [17], all of which is corre-
Individuals may develop limited mobility lated to the extent and severity of diabetic neu-
due to changes in connective and subcutaneous ropathy [18, 19]. Chronic hyperglycemia,
tissue leading to contractures and hammertoes. hypercholesterolemia, and hypertriglyceridemia
This may result in walking difficulties and are believed to be the main causes of these patho-
impairment of performing daily activities. As logical changes [1]. Severe demyelination with-
the disease progresses proximally, weakness out axonal loss is very rarely seen in DSPN and
may include the upper extremities, much like suggests concomitant neuropathy, due to an etiol-
the glove-and-­stocking distribution of sensory ogy other than diabetes. Many factors such as a
neuropathy. high BMI, inactivity, and myopathic changes in
186 K. S. Khan and H. Andersen

the muscles can contribute to the development of Myopathy is characterized by fibrosis and
motor impairment. atrophy of the muscle. This process causes loss
of energy reserves, decreased muscle quality,
and abnormal muscle structure, ultimately lead-
4 Neurogenic Muscle ing to muscle weakness and functional impair-
Weakness ments. Loss of muscle strength in myopathy is a
multifactorial process, which cannot be
In DSPN, muscle weakness is related to the explained by muscle atrophy or muscle disuse
severity of the neuropathy and develops due to alone. Other factors such as non-contractile tis-
degeneration of motor axons or entire motor sue infiltration and reduced muscle quality of
units leading to subsequent muscle fiber loss myocellular proteins may also contribute to
with changes in muscle morphology and atro- muscle weakness. Individuals with diabetic
phy [20–22]. In contrast to the clinical presenta- myopathy may appear clinically with normal
tion, in NCS, motor and sensory nerves are muscle mass, and nonetheless have lower muscle
impaired to a similar degree in the earlier phases strength due to lower quality of the muscles.
of DPN [23]. Muscular atrophy and hypoxia of the muscle
This indicates that more subtle clinical signs may contribute through impairment of mito-
of motor dysfunction are not primarily due to chondrial function. Altered mitochondrial func-
less severe pathology in motor nerves but rather tion contributes to insulin resistance of skeletal
is the consequence of the compensatory mecha- muscles due to lipid accumulation within myo-
nism named collateral re-innervation. Thereby, cytes, leading to a dysfunction of oxidative
denervated muscle fibers are re-innervated by enzymes. Abnormal mitochondrial morphology
neighboring motor nerves. However, as this re- in diabetes is also evident in tissues other than
innervation process becomes increasingly inad- muscle, including neurons. However, currently,
equate, accelerated loss of muscle strength there are no studies available in humans, and
occurs [21]. If denervated muscle fibers fail to most research was performed on diabetic mice.
acquire a new source of innervation, they atro- Another explanation for the development of dia-
phy and undergo apoptosis and ultimately necro- betic myopathy is recurrent administration of
sis with replacement by intramuscular fat subcutaneous insulin [15]. This is thought to
deposits and other non-contractile tissue. produce a chronic peripheral glucose overload
Furthermore, reduced levels of neurotrophic fac- causing a state of constant intracellular hyper-
tors in striated muscles of individuals with DPN glycemia contributing to the development of dia-
is related to muscle weakness and neuropathy. betic myopathy.
Thus, denervation of muscles in diabetes may be
further ­propagated by reduced levels of muscular
trophic factors and nerve growth factors [24]. 6 Diagnostic Approach

For the diagnosis of motor neuropathy, the cor-


5 Diabetic Myopathy nerstone is a clinical examination, including
manual muscle testing combined with electro-
More recently, there has been growing evidence physiological examinations. Most validated neu-
suggesting the existence of a diabetic myopathy ropathy scales assessing DSPN do not focus on
[15]. Diabetic myopathy is characterized by an motor involvement. In the Toronto criteria for
impairment in skeletal muscle structure, func- confirmed DSPN, early motor symptoms are not
tion, and metabolic capacity, which has been included in the standardized assessment for
described as a distinct process in diabetic indi- DSPN; therefore, motor involvement may be
viduals unrelated to DSPN. overlooked. Currently, the Toronto clinical neu-
Motor Neuropathy in Diabetes 187

ropathy score is the only clinical diabetic neu- In DSPN, a classical feature seen on ENG is
ropathy scale which includes symptoms of decreased amplitudes reflecting a loss of axons
muscle weakness [25]. combined with slowing in conduction velocity
reflecting abnormalities of the myelin sheath
[31]. Since typical diabetic neuropathy presents
7 Motor Nerve Dysfunction with features of both demyelination and axonal
loss, this is categorized as a mixed neuropathy.
Motor nerves can be evaluated using neurophysi- Abnormalities in sural nerve conduction is classi-
ological tests such as electroneurography (ENG) cally thought to be a sensitive measure to confirm
and electromyography (EMG). the diagnosis [32]. An unrecordable sural nerve
In individuals with typical bilateral, distal, action potential is a frequent finding in DSPN
symmetric polyneuropathy that present with both and can be interpreted as abnormal [33]. F-wave
signs and symptoms of DSPN, there is no need conduction velocity is an antidromic remnant of
for further ancillary tests to make the diagnoses the M-wave and is therefore only present in
at the level of possible and probable [26]. motor nerves. It is considered a valuable mea-
Ancillary investigations with NCS are needed to surement as it reflects pathology along the entire
confirm the diagnosis of typical DPSN, to diag- nerve and the anterior horn of the spinal cord.
nose subclinical DSPN, and to diagnose individ- Absent F-waves or prolonged F-wave latency are
uals with atypical features and presentations, or thought to be sensitive and reproducible mea-
when the diagnosis is uncertain [1, 26, 27]. sures of early DSPN [20, 34, 35]. This further
Confirmation of typical DSPN is usually only underscores the notion that motor nerves are
needed for research purposes [26, 28]. NCS can impaired to the same degree as the sensory nerves
also be used to quantify the severity of neuropa- in DSPN.
thy [2] and to measure disease progression [29]. Abnormalities in NCS do not differ between
Standard ENG examination includes an exami- type 1 and type 2 diabetes [36]; although the lit-
nation of both motor and sensory nerves. ENG erature in this area is sparse. Balducci et  al.
techniques, methods, materials, and equipment showed that while muscle strength was more
used may differ from examiner to examiner. decreased in patients with type 2 diabetes com-
However, there is some consensus on how an ENG pared to type 1 diabetes, there was no difference
examination should be performed and interpreted in NCS measurements [37]. Whether there are
in DSPN [30]. Generally, a NCS examination of differences in the pathogenesis of type 1 and type
the lower limb nerves, tibial, peroneal, and sural, is 2 DPN is unclear; therefore, nuances and differ-
sufficient. In screening for typical DSPN, a unilat- ences may exist in some studies. A histopatho-
eral examination is considered sufficient, provided logical study in rats showed that small-fiber
that the nerves are either abnormal or normal. The involvement might happen earlier in type 1 dia-
examination should be modified according to the betes [38]. However, this finding is not evident in
individual's signs and symptoms in order to accu- conventional NCS, as standard investigations
rately diagnose immune-mediated neuropathies, have limitations and only detect large-fiber
mononeuropathies, and radiculopathies (Table 1). abnormalities. Individuals with isolated small-­
With a standard ENG, the following NCS fiber or autonomic neuropathy may have a nor-
parameters are evaluated: Distal motor latency, mal ENG and a close to normal clinical
motor/sensory conduction velocity, compound examination, requiring ancillary diagnostics with
muscle action potential/sensory nerve action intraepidermal nerve fiber density (IENFD),
potential amplitude, and minimum F-wave quantitative sensory testing, and corneal confocal
latency [30]. microscopy [39–42].
188 K. S. Khan and H. Andersen

8 Electromyography 9 Muscle Strength in Diabetic


Distal Symmetric
Due to the length-dependent process in DSPN, atro- Polyneuropathy
phy and weakness of foot muscles are usually the
first clinical signs of motor involvement, followed Individuals with diabetes have reduced muscle
by weakness of ankle dorsiflexion resulting in foot strength when compared to healthy matched
drop. With further progression of the disease, hand controls [19, 37, 46]. This includes muscle
and forearm muscles may be involved. In the clini- groups in both upper and lower extremities. The
cal setting, EMG can be used to distinguish between lack of quantitative methods has disadvantaged
neurogenic weakness, due to axonal loss versus earlier studies when assessing muscle strength.
myogenic weakness from myopathy, and may also However, multiple recent studies have confirmed
aid as a supplementary investigation in suspicion of muscle weakness in diabetes combined with
a nerve root lesion. Preferably, EMG is performed findings documenting more pronounced muscle
in distal muscles using a concentric needle, as sur- weakness in individuals with DSPN.
face electrode EMG cannot quantify muscle qual- Furthermore, in DSPN, muscle weakness is pro-
ity. EMG should be performed in both a resting and gressive and related to the presence and espe-
an active muscle to evaluate motor unit action cially the severity of DSPN with a distal to
potential, recruitment, and interference patterns. As proximal distribution [18, 47]. Orlando et  al.
described above, loss of muscle strength in diabetes confirmed a correlation between motor and sen-
can be the consequence of both a primary muscular sory nerve conduction velocity and muscle
pathology (diabetic myopathy) [15], or secondary strength, and fatigability [48]. Several newer
pathology due to axonal loss leading to neurogenic studies have confirmed previous findings, indi-
weakness. Since the nerve damage found on ENG cating that the severity of DSPN is associated
is accompanied by neurogenic weakness found on with foot and ankle muscle weakness and a
EMG, performing an EMG is most often not neces- reduced range of motion of the lower extremities
sary in a clinical setting. Typical EMG findings in [49, 50]. In a recent publication, Orlando et al.
diabetic neurogenic weakness include; resting presented a review of the available literature sup-
fibrillation potentials, reduced recruitment patterns, porting the evidence for reduced muscle strength,
and broader, higher, polyphasic motor unit poten- altered muscular endurance, and muscle fiber
tials [29, 43]. These findings also correspond with composition in DSPN [14].
terminal axonal sprouting resulting in compensa- Increased muscle endurance has been reported
tory re-innervation. in long-term type 1 diabetes individuals [17, 51].
Skin biopsies provide an estimate on the num- However, other studies have shown opposite
ber of distal small nerve fiber segments; however, findings [14]. It has been speculated that changes
for obvious reasons, conventional EMG does not in muscle fiber type distributions could explain
provide a direct count of the number of motor the increased muscular endurance. Allen et  al.
axons. Novel methods such as MUNE can 2014 described a loss of fast-twitch motor units
­accurately estimate the entire number of motor and a decreased rate of torque development with
units within a muscle by using surface electrodes prolonged contractions in individuals with type 2
[44]. In a recent study, M-Scan MUNE evalua- diabetes and DSPN [52].
tions were performed in 21 individuals with Age-related sarcopenia may contribute to
DPN, 31 individuals with diabetes without DPN, decreased muscle size with increased intramus-
and 38 heathy controls. The authors were able to cular adipose tissue leading to alterations in glu-
detect axonal motor nerve damage when collat- cose disposal, propagating the development of
eral re-­innervation limited NCS changes, and the type 2 diabetes [53]. Some studies have shown
sensitivity of detecting motor involvement in that individuals with obesity and diabetes develop
DPN, was similar to that of sensory NCS [45]. muscle weakness and increased intramuscular
Motor Neuropathy in Diabetes 189

fat-accumulations, with hyperglycemia being the 10 Assessment of Muscle


main factor driving this mechanism [54, 55]. Strength
Park et  al., described that decreased muscle
strength was related to longer diabetes duration Muscle strength is defined as the maximum force
and poor glycemic control [19]. a muscle group or an individual muscle can gen-
In a recent meta-analysis, Gundmi et  al. erate [62]. Strength and force output is correlated
found that individuals with type 2 diabetes have to the cross-sectional area of muscle fibers
impaired hand function measured by grip and recruited to generate force and relies on motor
pinch strength when compared to healthy con- unit recruitment [63]. There is a large inter-­
trols [56]. Gutefeldt et  al. evaluated upper individual variation in muscle strength which can
extremity impairment in individuals with long- be explained primarily by differences in muscle
standing type 1 diabetes with and without DPN fiber types, training experience, age, sex, and
[57], and found that two-thirds of the individu- genetic capability.
als had one or more uni- or bilateral clinical Muscle strength can be evaluated by [64]:
signs of upper extremity impairment which cor-
related to a reduced grip force. However, in this 1. Functional tests
study, it remained unknown if the individuals 2. Manual muscle testing
had DPN and it would be speculative to attri- 3. Quantitative techniques.
bute upper extremity impairment to DPN. Lima
et  al. reported that individuals with DPN did Functional tests are easy to perform as they do
not have decreased hand force generation but not require any equipment, and therefore are suit-
impaired hand dexterity and handgrip force able for large-scale population-based studies, e.g.
control [58]. 6-min walk test (Fig.  1). Testing the ability to
Associations between diabetes and decreased
lung function have been reported [53]. Kabitz
et  al. provided one of the first descriptions of
impaired respiratory neuromuscular function
related to DSPN assessed by phrenic nerve stim-
ulation [59]. In a more recent study, Van Eetvelde
et al. described an association between the pres-
ence of DSPN and respiratory muscle strength in
individuals with type 2 diabetes compared to
healthy controls [60]. However, in their study,
results were not stratified according to DSPN;
therefore, no firm conclusion could be made
whether DSPN had a negative impact on respira-
tory muscle strength and function.
In a recent study by Venkataraman et al., indi-
viduals with DSPN had decreased activation of
the motor coordination and visual processing
regions, possibly caused by a reduced sensory
input from the foot [61]. They also found signs of
compensatory activity within the CNS, by greater
activation of motor coordination and sensory
facilitation regions, in individuals with DSPN
having lower muscle strength and decreased joint
mobility. Fig. 1  Six-minute walk test
190 K. S. Khan and H. Andersen

Dynamometry is a technique enabling testing


with both isometric and isokinetic movements
(Fig. 2). It provides quantitative information on a
linear scale reflecting static and dynamic strength
of all major muscle groups of the upper and lower
extremities. The main limitation of the test is that
the individual being tested has to be fully coop-
erative and factors such as lack of motivation,
pain, depression, and fatigue, may lead to sub-
maximal performance. Furthermore, dynamom-
etry is time-consuming, and equipment is
expensive, and usually only exists at exercise-­
science or neuromuscular specialist research
facilities.

11 Imaging of Muscular
Atrophy and Motor
Neuropathy

In DSPN with motor impairment, initial pre-


sentation is with atrophy of striated muscles
Fig. 2  Dynamometry enables testing of both isometric
and isokinetic movements occurring predominantly involving the smaller
muscles of the distal lower extremities. In later,
more advanced stages, proximal involvement
walk on heels and toes reflects the strength of the occurs. Muscle size can be quantitatively
ankle dorsal and plantar flexors, while rising determined with CT, USG, and MRI. USG is a
from a kneeling position reflects more proximal fast and inexpensive method, but has lower
muscle strength, including the hip and knee spatial resolution. CT is also a quick examina-
extensors, and flexors. Although easy to perform, tion, but has low tissue contrast and involves
functional tests have limitations. The exact rela- radiation exposure. A recent study by
tion to muscle strength is unclear, and the inabil- Henderson et  al. evaluated muscle size and
ity to perform these tests may be due to pain, muscle strength of the lower limbs by the use
obesity, postural instability, and other comorbidi- of USG in 15 individuals with diabetes and
ties unrelated to motor dysfunction. early DPN compared to healthy controls [65].
Manual muscle testing (MMT) is part of a stan- They found that intrinsic muscles of the lower
dard neurological examination and provides semi- limb were significantly smaller in the DPN
quantitative data expressed on an ordinal scale groups, which correlated to decreased muscle
reflecting muscle strength. MMT requires an expe- strength. Henderson suggested that USG is a
rienced examiner, and even then, manual testing is cheap and easily re-assessable method that is
insensitive compared to quantitative techniques. useful for the detection of progression of mus-
MMT has limited sensitivity in detecting mild to cle wasting in DPN.  In a previous study,
moderate symmetrical weakness and has adequate Severinsen et  al. showed that USG evaluation
sensitivity in cases with severe weakness. of small foot muscles identified considerable
Motor Neuropathy in Diabetes 191

atrophy which was related to the severity of individuals. It was suggested that prior to the
DPN based on NCS [66]. development of muscular atrophy, there are sub-
MRI has the highest resolution and can be stantial changes in the biochemical properties of
combined with quantitative techniques that the muscles [69].
enable a reliable estimation of muscle volume Almurdhi et  al. showed unexpectedly that
and the degree of fatty infiltration, extent of individuals with type 2 diabetes had proximal
extracellular edema, and changes in muscle rather than distal muscle weakness, related to the
microstructure. Furthermore, muscle energy severity of DSPN.  In their study, MRI revealed
properties have been studied using 31P-MRI reduced proximal muscle volume and weakness,
spectroscopy and show lower energy proper- whereas distal muscle weakness was unrelated to
ties in individuals with DSPN. MRI has several lower distal muscle volume [70]. The authors
disadvantages as part of a standardized diag- suggested that the finding was due to an increase
nostic tool, including high costs, lower avail- in distal intramuscular fat and non-contractile tis-
ability of equipment, and time-consuming sue, which was most pronounced in the plantar
scanning protocols. Individuals with pacemak- flexors. In a more recent MRI study of 40 indi-
ers, severe obesity, and claustrophobia are dif- viduals with type 2 diabetes and 20 healthy con-
ficult to evaluate by MRI.  In neuromuscular trols, our research group found that individuals
diseases, MRI studies of striated muscles have with DPN had decreased muscle strength and
focused on myopathies, whereas in DSPN, reduced muscle size compared to individuals
MRI has been applied for research purposes without DSPN, also being most pronounced at
only. the plantar flexors [71]. The muscle weakness
MRI enables quantification of lower limb observed was related mainly to muscle atrophy
muscular atrophy in individuals with DSPN [16]. and to a lesser extent, to decreased tissue quality,
Andersen et al. found a reduced volume of foot indicating a distal to proximal distribution as
muscles on MRI in neuropathic and non-­ expected in DSPN (Fig. 3).
neuropathic individuals, suggesting that foot To visualize peripheral nerves, USG has been
muscle atrophy can be an early phenomenon, used to evaluate the more superficial sural nerve
occurring even before clinical signs of DSPN are [72], but has limitations when trying to visualize
evident [16, 67]. In studies of individuals with more deeply situated mixed nerves in the lower
more severe DSPN, there was also a considerable extremities. MRI can be used to evaluate struc-
loss of striated muscle volume in the lower tural findings in larger mixed nerves and
extremities with a clear distal to proximal gradi- T2-weighted hyperintensities were more frequent
ent [18, 47]. In long-term follow-up studies, there in individuals with DSPN [73]. Diffusion-tensor-­
was evidence of progressive muscular atrophy imaging MR-Neurography enables the detection
and accelerated loss of muscle tissue and muscle of multifocal lesions of mixed peripheral nerves
strength [68]. in DSPN and enables differentiation between
In another study, individuals with DSPN had diabetic muscles with or without DSPN (Fig. 4)
impaired cellular metabolism with decreased [74]. In the future, higher resolution MR images
energy reserves in foot muscles, lower energy combined with automated post-processing may
reserves of inorganic phosphates, and a lower lead to more widespread use of MRI for monitor-
phosphocreatine ratio when compared to healthy ing DSPN.
192 K. S. Khan and H. Andersen

a HC –DPN +DPN
Calf Thigh Calf Thigh Calf Thigh

W
T1-weighted 4-point Dixon

b HC –DPN +DPN
Calf Thigh Calf Thigh Calf Thigh

TE = 7.6 ms
T2-mapping

TE = 129.2 ms

c HC –DPN +DPN
Calf Thigh Calf Thigh Calf Thigh

b = 0 s/mm3
DTI

b = 600 s/mm3

Fig. 3  Unprocessed MRI scans at calf and thigh levels in (TEs) (in milliseconds) illustrate the muscle signal loss
a healthy control (HC) participant, a participant without from the higher echo times. (c) Diffusion images obtained
diabetic polyneuropathy (-DPN), and a participant with with two different b-values, illustrate the muscle signal
DPN (+DPN). (a) Water (W) and fat (F) images obtained loss with higher b-values. (Image slices: brightness,
with four-point Dixon sequences were used to calculate +40%; contrast +20%). (Adapted with permission from
fat-fractions. (b) T2 images of two different echo times Radiology [71])
Motor Neuropathy in Diabetes 193

Fig. 4 (a1) Axial MRIs of the thigh, in healthy controls, of the shin, in a patient with diabetes without DPN. (c1)
(a2) Axial MRIs of the shin, in healthy controls, (a3) DTI Axial MRIs of the thigh, in a patient with diabetes and
trace images of the thigh, in healthy controls, (a4) DTI DPN, (c2) Axial MRIs of the shin, in a patient with diabe-
trace images of the shin, in healthy controls. (b1) Axial tes and DPN, (c3) DTI trace images of the thigh, in a
MRIs of the thigh, in a patient with diabetes without DPN, patient with diabetes and DPN, (c4) DTI trace images of
(b2) Axial MRIs of the shin, in a patient with diabetes the shin, in a patient with diabetes and DPN. (Adapted
without DPN, (b3) DTI trace images of the thigh, in a with permission from Diabetes [74])
patient with diabetes without DPN, (b4) DTI trace images
194 K. S. Khan and H. Andersen

12 Motor Function, Postural In a recent large cross-sectional study, we


Stability, and Falls found an increased number of reported falls
within the previous 12 months among individuals
Motor function and postural balance are highly with type 2 diabetes compared to healthy con-
dependent on muscle strength, and adequate trols, irrespective of the presence of DPN [85].
strength is required to prevent a fall [74]. Postural Individuals with type 2 diabetes that reported a
stability and proper ambulation rely on numerous fall had impaired postural stability, slower transi-
factors, including intact function of the visual, tional movements and decreased walking speed.
vestibular, somatosensory, and musculoskeletal However, to our surprise, muscle strength and
systems [75]. DSPN contributes to decreased scores of DPN did not differ between fallers and
stride length, impaired balance, poor coordina- non-fallers, although muscle strength correlated
tion, and unstable gait [76]. To avoid falls and to to the degree of postural instability and the degree
maintain postural stability during unexpected of DPN in both fallers and non-fallers. Therefore,
perturbations of movements or positions, quick physical tests and posturography, which are
compensatory muscle contractions are required. highly dependent on muscle strength, should be
These compensatory contractions may be incorporated in the design of future fall preven-
impaired in individuals with motor neuropathy tion screening programs.
due to muscle weakness and muscle atrophy of
the lower limbs. Several studies have reported a
higher frequency of falls in individuals with 13 Interventions
DSPN [8, 77, 78]. In a recent review of the cur-
rently available literature on DPN in relation to Currently, there are no disease-modifying treat-
falls, postural balance, and activities of daily liv- ments for diabetic motor neuropathy. Resistance-
ing, we found a robust body of evidence demon- and aerobic training has been shown to improve
strating lower postural stability and an increased glycemic and metabolic control, with a lower
frequency of falls in individuals with DSPN [79]. HbA1c, higher tissue insulin sensitivity, higher
However, only two studies have evaluated the HDL levels, lower plasma triglycerides, and
impact of DSPN on activities of daily living. reduced BMI, all in parallel with lower fat mass
Several studies have examined muscle strength and waist circumference [86–89]. Based on these
as a possible contributor to postural instability in findings, exercise is recommended for the pre-
diabetes. Simoneau et al. reported [80] an associa- vention of macro- and microvascular complica-
tion between postural instability and lower limb tions of diabetes.
muscle weakness in individuals with diabetes, but In individuals without DSPN, it is well estab-
found no relationship between muscle strength lished that exercise leads to gains in muscle
and severity of DSPN. Nardone et al. found that strength of the lower extremities by increasing not
postural instability was related to the impairment only the quality of the muscles, but also muscle
of sensory rather than motor nerve fibers [81] and mass. Until recently, individuals with DSPN were
this has been supported by Menz et al. [82] and advised to refrain from resistance and weight
Almurdhi et al. [83], who reported that peripheral bearing training due to concerns of foot ulcer-
loss of sensation was the main cause of postural ation. However, recent studies have disproved this
instability. Handsaker et al. found that individuals notion, deeming resistance and weight bearing
with DSPN had impaired strength generation at training to be safe in individuals with DSPN [90].
the ankle and knee while walking up and down Resistance training can prevent loss of stri-
stairs [84]. This finding was confirmed by ated muscles and improve muscle strength,
Richardson [7] and Allet [8] et  al., who argued insulin sensitivity, neural control in individuals
that both reduced proximal strength and distal with diabetes [91]. Further, resistance training
ankle proprioception predict postural instability, can induce hypertrophy of particularly type IIa
falls, and fall-related injuries. muscle fibers which enhances glucose utiliza-
Motor Neuropathy in Diabetes 195

tion and ATP generation. Castaneda et  al. week for 12  weeks [97]; however, DSPN was
showed that exercise improves glycemic control diagnosed based only on vibration perception
and neuromuscular function in individuals with- and was not confirmed by NCS. Handsaker et al.
out DPN [87]. studied nine individuals with DSPN diagnosed
Exercise training may be a preventive option to by two clinical tests [98] and found an improve-
delay the development of neuropathy in diabetes. ment in speed of ankle and knee strength genera-
In a prospective 4-year study by Balducci et al., tion during stair ascent and descent following
individuals with diabetes in the non-­intervention 1-h/week high-load dynamic resistance and iso-
group developed significantly more motor and metric exercises for 16 weeks.
sensory neuropathy compared to individuals per- In the studies by Kruse and Muller, exercise
forming long-term aerobic exercise training [92]. interventions did not lead to gains in muscle
Physical inactivity is associated with higher intra- strength; however, both studies included inter-
muscular adipose tissue in individuals with DPN ventions of low intensity bodyweight training. In
and type 2 diabetes [54]. Exercise combined with a randomized trial, Kruse and colleagues [98]
dietary interventions improves proximal cutane- examined the effects of bodyweight leg strength-
ous re-innervation and regenerative capacities in ening exercises and balance exercises over
individuals with pre-­ diabetes and neuropathy 12  months in 79 individuals with DSPN and
[93]. Kluding et al. recently reported that individ- found no difference in falls frequency, muscle
uals with DPN showed improved pain, neuropa- strength or balance between the groups following
thy symptoms, and intraepidermal nerve fiber the intervention. Mueller et al. compared weight-­
branching at proximal sites following aerobic and bearing versus non-weight-bearing exercises in
resistance exercise [94]. Singleton et  al. found individuals with DSPN and found no change in
improvements in IENFD in individuals with dia- muscle strength [99]. However, individuals exer-
betes without neuropathy following combined cised three times a week for 12  weeks, without
aerobic and resistance exercise [95]. In a random- resistance, which may explain the lack of an
ized study examining the effect of progressive effect on muscle strength.
resistance training we found improved muscle Progressive resistance training is the most
strength of the knee extensors and flexors and efficient way to gain muscle strength. Therefore,
motor function in individuals with type 2 diabetic it is surprising that previous studies have not
polyneuropathy at levels comparable with those included resistance throughout the exercise
seen in individuals with diabetes without DPN intervention. Resistance training can prevent
and healthy control individuals. No effects were muscle atrophy, improve neural control and pre-
observed in small nerve fiber function as reflected vent age-­ related sarcopenia with an improve-
in IENFD [96]. ment in gait speed, balance, functional mobility
Several studies have reported positive effects with a reduced risk of falls in healthy individuals
of training in individuals with DSPN; however, [81, 100, 101].
only a very limited number of studies have Several studies have reported improved motor
included quantitative evaluations of motor func- function measured by walking speed following
tion and/or muscle strength. In a small study of balance and gait training, bodyweight exercises,
eleven individuals with type 2 diabetes and DPN, aerobic exercise, and isokinetic dynamometry
Praet et al. found an increase in muscle strength combined with balance and treadmill exercises
following 10  weeks of progressive resistance [97, 99, 102, 103]. Furthermore, it has been
training combined with high-intensity interval shown that gait and balance improve following
training [12]. bodyweight exercises, balance training, or aero-
In a randomized trial, Allet et  al. found that bic exercise [104–107]. To our knowledge, no
individuals with and without DPN improved gait study has examined the effect of exercise in indi-
speed, balance, muscle strength and joint mobil- viduals with severe motor neuropathy. Further
ity, following gait and balance exercises twice a studies should focus on the severity of motor neu-
196 K. S. Khan and H. Andersen

ropathy by using quantitative techniques such as References


imaging and muscle biopsies to examine training
effects at the histopathological and biochemical 1. Pop-Busui R, Boulton AJM, Feldman EL, Bril V,
Freeman R, Malik RA, et  al. Diabetic neuropathy:
levels. a position statement by the American Diabetes
In individuals with DSPN and substantial mus- Association. Diabetes Care. 2017;40(1):136–54.
cle weakness, it remains unknown whether they [cited 2019 May 23]. http://www.ncbi.nlm.nih.gov/
respond to long-term resistance training consider- pubmed/27999003.
2. Dyck PJ, Carter RE, Litchy WJ.  Modeling nerve
ing their severe denervation and incomplete re- conduction criteria for diagnosis of diabetic poly-
innervation of striated muscles. Individuals with neuropathy. Muscle Nerve. 2011;44(3):340–5.
inherited neuropathies and severe weakness have [cited 2019 May 23]. http://www.ncbi.nlm.nih.gov/
been shown to improve their muscle strength fol- pubmed/21996793.
3. Dyck PJB, Overland CJ, Low PA, Litchy WJ, Davies
lowing training [108, 109]. JL, Dyck PJB, et  al. Signs and symptoms versus
Currently, there are no available evidence-­ nerve conduction studies to diagnose diabetic senso-
based treatment recommendations for individuals rimotor polyneuropathy: Cl vs. NPhys trial. Muscle
with motor involvement and diabetes. Several Nerve. 2010;42(2):157–64. [cited 2019 Dec 10].
http://doi.wiley.com/10.1002/mus.21661.
multimodal approaches, including lifestyle 4. Dyck PJ, Kratz KM, Karnes JL, Litchy WJ, Klein
changes with exercise, improved glycemic con- R, Pach JM, et al. The prevalence by staged severity
trol, and pharmacological treatments of comor- of various types of diabetic neuropathy, retinopathy,
bidities that accompany diabetes, have beneficial and nephropathy in a population-based cohort: the
Rochester Diabetic Neuropathy Study. Neurology.
effects and may alleviate symptoms of diabetes. 1993;43(4):817–24. http://www.ncbi.nlm.nih.gov/
Despite the severity and highly incapacitating pubmed/8469345.
nature of motor neuropathy there is a lack of 5. Suda EY, Matias AB, Bus SA, Sacco ICN, et  al.
large detailed quantitative studies on motor affec- Impact of diabetic neuropathy severity on foot clear-
ance complexity and variability during walking. Gait
tion and the assessment of treatment options. Posture. 2019;74:194–9. https://doi.org/10.1016/j.
gaitpost.2019.09.014.
6. Martinelli AR, Mantovani AM, Nozabieli AJL,
14 Conclusions Ferreira DMA, Barela JA, de Camargo MR, et  al.
Muscle strength and ankle mobility for the gait
parameters in diabetic neuropathies. Foot (Edinb).
• Motor neuropathy presents with distal sym- 2013;23(1):17–21.
metrical impairment always in combination 7. Richardson JK, Demott T, Allet L, Kim H, Ashton-­
with sensory disturbances. Miller JA.  Hip strength: ankle proprioceptive
threshold ratio predicts falls and injury in diabetic
• Motor neuropathy presents in a length-­ neuropathy. Muscle Nerve. 2014;50(3):437–42.
dependent pattern with a clear proximal-distal 8. Allet L, Kim H, Ashton-Miller J, De Mott T,
gradient. Richardson JK.  Step length after discrete pertur-
• The diagnosis of motor neuropathy is made bation predicts accidental falls and fall-related
injury in elderly people with a range of peripheral
based on a combined assessment of motor neuropathy. J Diabetes Complications. 2014;28(1
nerve function and muscle strength. CC-Injuries):79–84. https://www.cochranelibrary.
• Motor dysfunction is clinically evident in only com/central/doi/10.1002/central/CN-­01038023/full.
a small proportion of individuals with 9. Cavanagh PR, Derr JA, Ulbrecht JS, Maser RE,
Orchard TJ. Problems with gait and posture in neu-
diabetes. ropathic patients with insulin-dependent diabetes
• Motor dysfunction may develop not only due mellitus. Diabet Med. 1992;9(5):469–74.
to motor neuropathy but also as a consequence 10. Onodera AN, Gomes AA, Pripas D, Mezzarane
of lower muscle quality due to diabetic RA, Sacco ICN.  Lower limb electromygraphy and
kinematics of neuropathic diabetic patients during
myopathy. real-life activities: stair negotiation. Muscle Nerve.
• Resistance training is a hitherto underutilized 2011;44(2):269–77. http://www.embase.com/
non-pharmacological treatment modality that search/results?subaction=viewrecord&from=export
may be useful for individuals with diabetic &id=L362173554.
polyneuropathy and motor dysfunction.
Motor Neuropathy in Diabetes 197

11. Andersen H, Mogensen PM.  Disordered mobil- to neuropathy and muscle strength. J Diabetes.
ity of large joints in association with neuropathy in 2014;6(5):462–71.
patients with long-standing insulin-dependent diabe- 23. Meijer JWK, Bosma E, Lefrandt JD, Links TP,
tes mellitus. Diabet Med. 1997;14(3):221–7. Smith AJ, Steward R.  Symptom scoring systems
12. Praet SFE, Jonkers RAM, Schep G, Stehouwer to diagnose distal polyneuropathy in diabetes: the
CDA, Kuipers H, Keizer HA, et al. Long-standing, diabetic neuropathy symptom score. Diabet Med.
insulin-treated type 2 diabetes patients with com- 2002;19:962–5.
plications respond well to short-term resistance 24. Andreassen CS, Jakobsen J, Flyvbjerg A, Andersen
and interval exercise training. Eur J Endocrinol. H.  Expression of neurotrophic factors in diabetic
2008;158(2):163–72. [cited 2019 Jul 9]. http://www. muscle-relation to neuropathy and muscle strength.
ncbi.nlm.nih.gov/pubmed/18230822. Brain. 2009;132(10):2724–33.
13. Balducci S, Zanuso S, Massarini M, Corigliano G, 25. Bril V, Perkins BA.  Validation of the Toronto
Nicolucci A, Missori S, et  al. The Italian Diabetes Clinical Scoring System for diabetic polyneuropa-
and Exercise Study (IDES): design and methods for thy. Diabetes Care. 2002;25(11):2048–52. [cited
a prospective Italian multicentre trial of intensive 2019 May 24]. http://www.ncbi.nlm.nih.gov/
lifestyle intervention in people with type 2 diabetes pubmed/12401755.
and the metabolic syndrome. Nutr Metab Cardiovasc 26. Tesfaye S, Boulton AJM, Dyck PJ, Freeman R,
Dis. 2008;18(9):585–95. [cited 2019 Jul 17]. http:// Horowitz M, Kempler P, et  al. Diabetic neuropa-
www.ncbi.nlm.nih.gov/pubmed/18061415. thies: update on definitions, diagnostic criteria, esti-
14. Orlando G, Balducci S, Bazzucchi I, Pugliese G, mation of severity, and treatments. Diabetes Care.
Sacchetti M.  Neuromuscular dysfunction in type 2010;33(10):2285–93. [cited 2019 May 23]. http://
2 diabetes: underlying mechanisms and effect www.ncbi.nlm.nih.gov/pubmed/20876709.
of resistance training. Diabetes Metab Res Rev. 27. Callaghan BC, Xia R, Reynolds E, Banerjee
2016;32(1):40–50. [cited 2019 Jul 9] http://doi. M, Rothberg AE, Burant CF, et  al. Association
wiley.com/10.1002/dmrr.2658. between metabolic syndrome components and
15. Monaco CMF, Perry CGR, Hawke TJ.  Diabetic polyneuropathy in an obese population. JAMA
myopathy. Curr Opin Neurol. 2017;30(5):545–52. Neurol. 2016;73(12):1468. [cited 2020 May
[cited 2019 Aug 14]. http://insights.ovid.com/ 10]. https://www.ncbi.nlm.nih.gov/pmc/articles/
crossref?an=00019052-­201710000-­00016. PMC5217829/#R30.
16. Andersen H, Gadeberg PC, Brock B, Jakobsen 28. Dyck PJ, Albers JW, Andersen H, Arezzo JC,
J.  Muscular atrophy in diabetic neuropathy: a ste- Biessels G-J, Bril V, et  al. Diabetic polyneuropa-
reological magnetic resonance imaging study. thies: update on research definition, diagnostic cri-
Diabetologia. 1997;40(9):1062–9. teria and estimation of severity. Diabetes Metab Res
17. Andersen H.  Muscular endurance in long-term Rev. 2011;27(7):620–8.
IDDM patients. Diabetes Care. 1998;21(4):604–9. 29. Gregersen G. Latency time, maximal amplitude and
http://www.embase.com/search/results?subaction=v electromyography in diabetic patients. Acta Med
iewrecord&from=export&id=L28172942. Scand. 1968;183(1–2):55–60.
18. Andersen H, Jacobsen J, Andreassen CS.  A pro- 30. Tankisi H, Pugdahl KBS, Andersen HF-FA, Tankisi
gressive late complication in diabetic distal H, Pugdahl K, Beniczky S, Andersen H, et  al.
symmetric polyneuropathy. Muscle Weakness. Evidence-based recommendations for examination
2006;21(2):262–8. and diagnostic strategies of polyneuropathy electro-
19. Park SW, Goodpaster BH, Strotmeyer ES, de diagnosis. Clin Neurophysiol Pract. 2019;4:214–22.
Rekeneire N, Harris TB, Schwartz AV, et  al. [cited 2019 Dec 14]. https://www.sciencedirect.com/
Decreased muscle strength and quality in older adults science/article/pii/S2467981X19300381.
with type 2 diabetes: the health, aging, and body 31. Weisman A, Bril V, Ngo M, Lovblom LE, Halpern
composition study. Diabetes. 2006;55(6):1813–8. EM, Orszag A, et  al. Identification and prediction
20. Andersen H, Stålberg E, Falck B.  F-wave latency, of diabetic sensorimotor polyneuropathy using indi-
the most sensitive nerve conduction parameter vidual and simple combinations of nerve conduction
in patients with diabetes mellitus. Muscle Nerve. study parameters. PLoS One. 2013;8(3):e58783.
1997;20(10):1296–302. 32. Albers JW, Brown MB, Sima AA, Greene DA. Nerve
21. Andersen H, Stålberg E, Gjerstad MD, Jakobsen conduction measures in mild diabetic neuropathy
J. Association of muscle strength and electrophysi- in the Early Diabetes Intervention Trial: the effects
ological measures of reinnervation in diabetic neu- of age, sex, type of diabetes, disease duration, and
ropathy. Muscle Nerve. 1998;21(12):1647–54. anthropometric factors. Tolrestat Study Group for
[cited 2019 Dec 7]. http://www.ncbi.nlm.nih.gov/ the Early Diabetes Intervention Trial. Neurology.
pubmed/9843064. 1996;46(1):85–91.
22. Andreassen CS, Jensen JM, Jakobsen J, Ulhøj BP, 33. Dyck PJ, Karnes JL, Daube J, O’Brien P, Service
Andersen H.  Striated muscle fiber size, composi- FJ.  Clinical and neuropathological criteria for the
tion, and capillary density in diabetes in relation diagnosis and staging of diabetic polyneuropathy.
198 K. S. Khan and H. Andersen

Brain. 1985;108(4):861–80. https://doi.org/10.1093/ 46. Lord SR, Caplan GA, Colagiuri R, Colagiuri S,
brain/108.4.861. Ward JA.  Sensori-motor function in older persons
34. Kohara N, Kimura J, Kaji R, Goto Y, Ishii J, with diabetes. Diabet Med. 1993;10(7):614–8.
Takiguchi M, et al. F-wave latency serves as the most 47. Andersen H, Poulsen PL, Mogensen CE, Jakobsen
reproducible measure in nerve conduction studies J.  Isokinetic muscle strength in long-term IDDM
of diabetic polyneuropathy: multicentre analysis in patients in relation to diabetic complications.
healthy subjects and patients with diabetic polyneu- Diabetes. 1996;45(4):440–5. [cited 2019 Dec 13].
ropathy. Diabetologia. 2000;43(7):915–21. http://www.ncbi.nlm.nih.gov/pubmed/8603765.
35. Pan H, Lin J, Chen N, Jian F, Zhang Z, Ding Z, et al. 48. Orlando G, Balducci S, Bazzucchi I, Pugliese G,
Normative data of F-wave measures in China. Clin Sacchetti M. The impact of type 1 diabetes and dia-
Neurophysiol. 2013;124(1):183–9. betic polyneuropathy on muscle strength and fatiga-
36. Hendriksen PH, Oey PL, Wieneke GH, Bravenboer bility. Acta Diabetol. 2017;54(6):543–50.
B, Banga JD.  Subclinical diabetic neuropathy: 49. Nomura T, Ishiguro T, Ohira M, Ikeda Y.  Diabetic
similarities between electrophysiological results polyneuropathy is a risk factor for decline of lower
of patients with Type 1 (insulin-dependent) and extremity strength in patients with type 2 diabetes. J
Type 2 (non-insulin-dependent) diabetes melli- Diabetes Investig. 2018;9(1):186–92.
tus. Diabetologia. 1992;35(7):690–5. https://doi. 50. Bursac SN, Jandric S, Talic G. Influence of diabetic
org/10.1007/BF00400264. distal symmetric polyneuropathy on the perfor-
37. Balducci S, Sacchetti M, Orlando G, Salvi L, mance of the musculoskeletal system of lower leg
Pugliese L, Salerno G, et  al. Correlates of muscle and foot. Med Arch. 2019;73(4):262–7.
strength in diabetes: the study on the assessment of 51. IJzerman TH, Schaper NC, Melai T, Meijer K,
determinants of muscle and bone strength abnormal- Willems PJB, Savelberg HHCM.  Lower extremity
ities in diabetes (SAMBA). Nutr Metab Cardiovasc muscle strength is reduced in people with type 2 dia-
Dis. 2014;24(1):18–26. betes, with and without polyneuropathy, and is asso-
38. Kamiya H, Murakawa Y, Zhang W, Sima ciated with impaired mobility and reduced quality
AAF. Unmyelinated fiber sensory neuropathy differs of life. Diabetes Res Clin Pract. 2012;95(3):345–51.
in type 1 and type 2 diabetes. Diabetes Metab Res 52. Allen MD, Major B, Kimpinski K, Doherty TJ, Rice
Rev. 2005;21(5):448–58. CL.  Skeletal muscle morphology and contractile
39. Singleton JR, Bixby B, Russell JW, Feldman function in relation to muscle denervation in diabetic
EL, Peltier A, Goldstein J, et  al. The Utah Early neuropathy. J Appl Physiol. 2014;116(5):545–52.
Neuropathy Scale: a sensitive clinical scale for early 53. Mesinovic J, Zengin A, De Courten B, Ebeling PR,
sensory predominant neuropathy. J Peripher Nerv Scott D.  Sarcopenia and type 2 diabetes mellitus:
Syst. 2008;13(3):218–27. [cited 2019 May 24]. a bidirectional relationship. Diabetes Metab Syndr
http://www.ncbi.nlm.nih.gov/pubmed/18844788. Obes. 2019;12:1057–72.
40. Devigili G, Tugnoli V, Penza P, Camozzi F, Lombardi 54. Tuttle LJ, Sinacore DR, Cade WT, Mueller
R, Melli G, et  al. The diagnostic criteria for small MJ.  Lower physical activity is associated with
fibre neuropathy: from symptoms to neuropathol- higher intermuscular adipose tissue in people with
ogy. Brain. 2008;131(7):1912. [cited 2020 May type 2 diabetes and peripheral neuropathy. Phys
14]. https://www.ncbi.nlm.nih.gov/pmc/articles/ Ther. 2011;91(6):923–30. [cited 2019 Jul 9]. http://
PMC2442424/. www.ncbi.nlm.nih.gov/pubmed/21474636.
41. Sivaskandarajah GA, Halpern EM, Lovblom LE, 55. Bittel AJ, Bittel DC, Tuttle LJ, Strube MJ, Mueller
Weisman A, Orlov S, Bril V, et al. Structure-function MJ, Cade WT, et  al. Explanators of Sarcopenia in
relationship between corneal nerves and conven- individuals with diabesity: a cross-sectional analy-
tional small-fiber tests in type 1 diabetes. Diabetes sis. J Geriatr Phys Ther. 2017;40(2):86–94.
Care. 2013;36(9):2748–55. 56. Gundmi S, Maiya AG, Bhat AK, Ravishankar
42. Andersson C, Guttorp P, Särkkä A.  Discovering N, Hande MH, Rajagopal KV.  Hand dysfunc-
early diabetic neuropathy from epidermal nerve fiber tion in type 2 diabetes mellitus: systematic review
patterns. Stat Med. 2016;35(24):4427–42. with meta-analysis. Ann Phys Rehabil Med.
43. Braune HJ.  Early detection of diabetic neu- 2018;61(2):99–104.
ropathy: a neurophysiological study on 100 57. Gutefeldt K, Lundstedt S, Thyberg ISMM,
patients. Electromyogr Clin Neurophysiol. Bachrach-Lindström M, Arnqvist HJ, Spångeus
1997;37(7):399–407. A.  Clinical examination and self-reported upper
44. Jacobsen AB, Bostock H, Tankisi H.  CMAP scan extremity impairments in patients with long-­
MUNE (MScan)—a novel motor unit number esti- standing type 1 diabetes mellitus. J Diabetes Res.
mation (MUNE) method. J Vis Exp. 2018;(136). 2020;2020:4172635.
45. Kristensen AG, Khan KS, Bostock H, Khan BS, 58. Lima KCA, Borges LS, Hatanaka E, Rolim LC, de
Gylfadottir S, Andersen H, et  al. MScanFit motor Freitas PB. Grip force control and hand dexterity are
unit number estimation and muscle velocity recov- impaired in individuals with diabetic peripheral neu-
ery cycle recordings in diabetic polyneuropathy. ropathy. Neurosci Lett. 2017;659:54–9.
Clin Neurophysiol Pract. 2020;131(11):2591–9.
Motor Neuropathy in Diabetes 199

59. Kabitz H-J, Sonntag F, Walker D, Schwoerer A, 72. Kim K-H, Yoo J-Y, You B-C.  Ultrasonographic
Walterspacher S, Kaufmann S, et al. Diabetic poly- evaluation of sural nerve for nerve conduction study.
neuropathy is associated with respiratory muscle Ann Rehabil Med. 2014;38(1):46–51.
impairment in type 2 diabetes. Diabetologia. 73. Pham M, Oikonomou D, Bäumer P, Bierhaus A,
2008;51(1):191–7. Heiland S, Humpert PM, et al. Proximal neuropathic
60. Van Eetvelde BLM, Cambier D, Vanden Wyngaert lesions in distal symmetric diabetic polyneuropathy:
K, Celie B, Calders P.  The influence of clini- findings of high-resolution magnetic resonance neu-
cally diagnosed neuropathy on respiratory muscle rography. Diabetes Care. 2011;34(3):721–3.
strength in type 2 diabetes mellitus. J Diabetes Res. 74. Vaeggemose M, Pham M, Ringgaard S, Tankisi H,
2018;2018:8065938. Ejskjaer N, Heiland S, et  al. Magnetic resonance
61. Venkataraman K, Pun V, Mohamed AZ, Luo M, neurography visualizes abnormalities in sciatic and
Wong C, Zong F, et al. Altered motor and motor per- tibial nerves in patients with type 1 diabetes and neu-
ceptual cognitive imagery task-related activation in ropathy. Diabetes. 2017;66(7):1779–88.
diabetic peripheral neuropathy: insights from func- 75. Hewston P, Deshpande N. Falls and balance impair-
tional MRI. Diabetes Care. 2019;42(10):2004–7. ments in older adults with type 2 diabetes: think-
62. Kell RT, Bell G, Quinney A.  Musculoskeletal fit- ing beyond diabetic peripheral neuropathy. Can J
ness, health outcomes and quality of life. Sport Med. Diabetes. 2016;40(1):6–9. [cited 2017 Jan 9]. http://
2001;31(12):863–73. [cited 2019 Dec 19]. http:// www.embase.com/search/results?subaction=viewre
www.ncbi.nlm.nih.gov/pubmed/11665913. cord&from=export&id=L607755496.
63. Wakeling JM, Lee SSM, Arnold AS, de Boef Miara 76. Brown SJ, Handsaker JC, Bowling FL, Boulton AJM,
M, Biewener AA. A muscle’s force depends on the Reeves ND. Diabetic peripheral neuropathy compro-
recruitment patterns of its fibers. Ann Biomed Eng. mises balance during daily activities. Diabetes Care.
2012;40(8):1708–20. [cited 2019 Dec 13]. http:// 2015;38(6):1116–22. [cited 2019 May 23]. http://
www.ncbi.nlm.nih.gov/pubmed/22350666. www.embase.com/search/results?subaction=viewre
64. Andersen H, Nielsen S, Mogensen CE, Jakobsen cord&from=export&id=L609629141.
J.  Muscle strength in type 2 diabetes. Diabetes. 77. Vaz MM, Costa GC, Reis JG, Junior WM,
2004;53(6):1543–8. [cited 2019 Jul 23]. http://diabe- Albuquerque de Paula FJ, Abreu DC. Postural control
tes.diabetesjournals.org/content/53/6/1543.abstract. and functional strength in patients with type 2 diabe-
65. Henderson AD, Johnson AW, Rasmussen LG, Peine tes mellitus with and without peripheral neuropathy.
WP, Symons SH, Scoresby KA, et  al. Early-stage Arch Phys Med Rehabil. 2013;94(12):2465–70.
diabetic neuropathy reduces foot strength and intrin- 78. Richardson JK, Eckner JT, Allet L, Kim H, Ashton-­
sic but not extrinsic foot muscle size. J Diabetes Res. Miller JA.  Complex and simple clinical reaction
2020;2020:9536362. times are associated with gait, balance, and major
66. Severinsen K. Atrophy of foot muscles in diabetic. fall injury in older subjects with diabetic peripheral
Diabetes Care. 2007;30(12):3053–7. neuropathy. Am J Phys Med Rehabil. 2017;96(1):8–
67. Severinsen K, Andersen H. Evaluation of atrophy of 16. http://www.embase.com/search/results?subactio
foot muscles in diabetic neuropathy—a comparative n=viewrecord&from=export&id=L617662636.
study of nerve conduction studies and ultrasonogra- 79. Khan KS, Andersen H. The impact of diabetic neu-
phy. Clin Neurophysiol. 2007;118(10):2172–5. ropathy on activities of daily living, postural balance
68. Andreassen CS, Jakobsen J, Ringgaard S, Ejskjaer and risk of falls—a systematic review. J Diabetes Sci
N, Andersen H. Accelerated atrophy of lower leg and Technol. 2022;16(2):289–94.
foot muscles-a follow-up study of long-term diabetic 80. Simoneau GG, Ulbrecht JS, Derr JA, Becker MB,
polyneuropathy using magnetic resonance imaging Cavanagh PR.  Postural instability in patients
(MRI). Diabetologia. 2009;52(6):1182–91. with diabetic sensory neuropathy. Diabetes Care.
69. Greenman RL, Panasyuk S, Wang X, Lyons TE, 1994;17(12):1411–21. [cited 2019 Jul 23]. https://
Dinh T, Longoria L, et al. Early changes in the skin doi.org/10.2337/diacare.17.12.1411.
microcirculation and muscle metabolism of the dia- 81. Abe T, De Hoyos DV, Pollock ML, Garzarella
betic foot. Lancet. 2005;366(9498):1711–7. L.  Time course for strength and muscle thick-
70. Almurdhi MM, Reeves ND, Bowling FL, Boulton ness changes following upper and lower body
AJM, Jeziorska M, Malik RA.  Reduced lower-­ resistance training in men and women. Eur J Appl
limb muscle strength and volume in patients with Physiol Occup Physiol. 2000;81(3):0174. [cited
type 2 diabetes in relation to neuropathy, intra- 2019 Dec 23]. http://www.ncbi.nlm.nih.gov/
muscular fat, and vitamin D levels. Diabetes Care. pubmed/10638374.
2016;39(3):441–7. 82. Menz HB, Lord SR, St George R, Fitzpatrick RC,
71. Stouge A, Khan KS, Kristensen AG, Tankisi H, George RS, Fitzpatrick RC, et  al. Walking stabil-
Schlaffke L, Froeling M, et  al. MRI of skeletal ity and sensorimotor function in older people with
muscles in participants with type 2 diabetes with diabetic peripheral neuropathy. Arch Phys Med
or without diabetic polyneuropathy. Radiology. Rehabil. 2004;85(2):245–52. [cited 2019 May 23].
2020;297(3):608–19. http://www.ncbi.nlm.nih.gov/pubmed/14966709.
200 K. S. Khan and H. Andersen

83. Almurdhi MM, Brown SJ, Bowling FL, Boulton 2006;20(4):216–23. [cited 2019 Jul 17]. https://
AJM, Jeziorska M, Malik RA, et al. Altered walking www.sciencedirect.com/science/article/abs/pii/S105
strategy and increased unsteadiness in participants 6872705000905?via%3Dihub.
with impaired glucose tolerance and Type 2 diabetes 93. Smith AG, Russell J, Feldman EL, Goldstein
relates to small-fibre neuropathy but not vitamin D J, Peltier A, Smith S, et  al. Lifestyle interven-
deficiency. Diabet Med. 2017;34(6):839–45. tion for pre-diabetic neuropathy. Diabetes Care.
84. Handsaker JC, Brown SJ, Bowling FL, Cooper G, 2006;29(6):1294–9.
Maganaris CN, Boulton AJM, et  al. Contributory 94. Kluding PM, Pasnoor M, Singh R, Jernigan S,
factors to unsteadiness during walking up and down Farmer K, Rucker J, et  al. The effect of exercise
stairs in patients with diabetic peripheral neuropathy. on neuropathic symptoms, nerve function, and
Diabetes Care. 2014;37(11):3047–53. [cited 2019 cutaneous innervation in people with diabetic
Aug 4]. https://doi.org/10.2337/dc14-­0955. peripheral neuropathy. J Diabetes Complications.
85. Khan KS, Pop-Busui R, Devantier L, Kristensen 2012;26(5):424–9. [cited 2019 Jul 9]. http://www.
AG, Tankisi H, Dalgas U, et  al. Falls in individu- ncbi.nlm.nih.gov/pubmed/22717465.
als with type 2 diabetes; a cross-sectional study on 95. Singleton JR, Marcus RL, Jackson JE, Lessard M,
the impact of motor dysfunction, postural insta- Graham TE, Smith AG.  Exercise increases cutane-
bility and diabetic polyneuropathy. Diabet Med. ous nerve density in diabetic patients without neu-
2020;38(9):e14470. ropathy. Ann Clin Transl Neurol. 2014;1(10):844–9.
86. Church TS, Blair SN, Cocreham S, Johannsen [cited 2019 Jul 17]. http://www.ncbi.nlm.nih.gov/
N, Johnson W, Kramer K, et  al. Effects of aero- pubmed/25493275.
bic and resistance training on hemoglobin A1c 96. Khan KS, Overgaard K, Tankisi H, Karlsson P,
levels in patients with type 2 diabetes. JAMA. Devantier L, Gregersen S, et al. Effects of progressive
2010;304(20):2253. [cited 2019 Dec 20]. http:// resistance training in individuals with type 2 diabetic
www.ncbi.nlm.nih.gov/pubmed/21098771. polyneuropathy: a randomised assessor-blinded con-
87. Castaneda C, Layne JE, Munoz-Orians L, Gordon trolled trial. Diabetologia. 2022;65(4):620–31.
PL, Walsmith J, Foldvari M, et  al. A randomized 97. Allet L, Armand S, de Bie RA, Golay A, Monnin D,
controlled trial of resistance exercise training to Aminian K, et  al. The gait and balance of patients
improve glycemic control in older adults with type with diabetes can be improved: a randomised con-
2 diabetes. Diabetes Care. 2002;25(12):2335–41. trolled trial. Diabetologia. 2010;53(3):458–66.
[cited 2019 Aug 17]. http://www.ncbi.nlm.nih.gov/ [cited 2019 Jul 6]. https://www.ncbi.nlm.nih.gov/
pubmed/12453982. pmc/articles/PMC2815802/.
88. Colberg SR, Sigal RJ, Fernhall B, Regensteiner JG, 98. Robin LK, Joseph WL, Richard WM. Fall and
Blissmer BJ, Rubin RR, et  al. Exercise and type 2 balance outcomes after an intervention to pro-
diabetes: the American College of Sports Medicine mote leg strength, balance, and walking in peo-
and the American Diabetes Association: joint posi- ple with diabetic peripheral neuropathy: “Feet
tion statement. Diabetes Care. 2010;33(12):e147. First” Randomized Controlled Trial. https://doi.
[cited 2019 Aug 23]. https://www.ncbi.nlm.nih.gov/ org/10.2522/ptj.20090362.
pmc/articles/PMC2992225/. 99. Mueller MJ, Tuttle LJ, Lemaster JW, Strube MJ,
89. Dagogo-Jack S, Egbuonu N, Edeoga C.  Principles McGill JB, Hastings MK, et  al. Weight-bearing
and practice of nonpharmacological interventions versus nonweight-bearing exercise for persons
to reduce cardiometabolic risk. Med Princ Pract. with diabetes and peripheral neuropathy: a ran-
2010;19(3):167–75. [cited 2019 Dec 10]. http:// domized controlled trial. Arch Phys Med Rehabil.
www.ncbi.nlm.nih.gov/pubmed/20357497. 2013;94(5):829–38. [cited 2019 Jul 9]. https://doi.
90. LeMaster JW, Mueller MJ, Reiber GE, Mehr DR, org/10.1016/j.apmr.2012.12.015.
Madsen RW, Conn VS.  Effect of weight-bearing 100. Mcleod JC, Stokes T, Phillips SM. Resistance exer-
activity on foot ulcer incidence in people with dia- cise training as a primary countermeasure to age-­
betic peripheral neuropathy: feet first randomized related chronic disease. Front Physiol. 2019;10:645.
controlled trial. Phys Ther. 2008;88(11):1385–98. 101. Cadore EL, Rodríguez-Mañas L, Sinclair A,
[cited 2019 Aug 18]. http://www.ncbi.nlm.nih.gov/ Izquierdo M. Effects of different exercise interven-
pubmed/18801859. tions on risk of falls, gait ability, and balance in
91. Sigal RJ, Kenny GP, Wasserman DH, Castaneda-­ physically frail older adults: a systematic review.
Sceppa C.  Physical activity/exercise and type 2 Rejuvenation Res. 2013;16(2):105–14.
diabetes. Diabetes Spectr. 2005;18(1):2518–39. 102. Morrison S, Colberg SR, Parson HK, Vinik
[cited 2019 Aug 31]. http://www.ncbi.nlm.nih.gov/ AI.  Exercise improves gait, reaction time and
pubmed/15451933. postural stability in older adults with type 2 dia-
92. Balducci S, Iacobellis G, Parisi L, Di Biase N, betes and neuropathy. J Diabetes Complications.
Calandriello E, Leonetti F, et  al. Exercise train- 2014;28(5):715–22. [cited 2019 Jul 9]. http://www.
ing can modify the natural history of diabetic embase.com/search/results?subaction=viewrecord&
peripheral neuropathy. J Diabetes Complications. from=export&id=L53187427.
Motor Neuropathy in Diabetes 201

103. Taveggia G, Villafañe JH, Vavassori F, Lecchi C, training: a single blind, randomized controlled
Borboni A, Negrini S, et  al. Multimodal treat- trial. Am J Phys Med Rehabil. 2016;95(7):516–24.
ment of distal sensorimotor polyneuropathy in [cited 2019 Jul 9]. http://www.ncbi.nlm.nih.gov/
diabetic patients: a randomized clinical trial. J pubmed/26745226.
Manipulative Physiol Ther. 2014;37(4):242–52. 107. Song CH, Petrofsky JS, Lee SW, Lee KJ, Yim
[cited 2016 Oct 21]. http://www.ncbi.nlm.nih.gov/ JE.  Effects of an exercise program on balance
pubmed/24656867. and trunk proprioception in older adults with
104. Richardson JK, Sandman D, Vela S. A focused exer- diabetic neuropathies. Diabetes Technol Ther.
cise regimen improves clinical measures of balance 2011;13(8):803–11. http://www.ncbi.nlm.nih.gov/
in patients with peripheral neuropathy. Arch Phys pubmed/21561371.
Med Rehabil. 2001;82(2):205–9. http://www.ncbi. 108. Chetlin RD, Gutmann L, Tarnopolsky MA, Ullrich
nlm.nih.gov/pubmed/11239311. IH, Yeater RA. Resistance training exercise and cre-
105. Akbari M, Jafari H, Moshashaee A, Forugh B.  Do atine in patients with Charcot-Marie-Tooth disease.
diabetic neuropathy patients benefit from balance Muscle Nerve. 2004;30(1):69–76.
training? J Rehabil Res Dev. 2012;49(2):333–8. 109. Burns J, Sman AD, Cornett KMD, Wojciechowski E,
[cited 2019 Jul 9]. http://www.ncbi.nlm.nih.gov/ Walker T, Menezes MP, et al. Safety and efficacy of
pubmed/22773533. progressive resistance exercise for Charcot-Marie-­
106. Dixit S, Maiya A, Shastry BA, Guddattu V. Analysis Tooth disease in children: a randomised, double-­
of postural control during quiet standing in a blind, sham-controlled trial. Lancet Child Adolesc
population with diabetic peripheral neuropathy Heal. 2017;1(2):106–13.
undergoing moderate intensity aerobic exercise
Cardiovascular Autonomic
Neuropathy

Lynn Ang and Rodica Pop-Busui

1 Cardiovascular Autonomic 2 Epidemiology


Neuropathy
The reported prevalence of CAN varies depend-
Autonomic neuropathies affect the autonomic ing on the population(s) studied (e.g., type 1 dia-
neurons (parasympathetic, sympathetic, or both) betes versus type 2 diabetes, intervention trials
and manifest as a variety of condition-specific versus epidemiological observations, earlier ver-
symptoms and signs involving the cardiovascu- sus more contemporary cohorts), the available
lar, gastrointestinal, genitourinary, and sudomo- outcome measures reported, and/or the diagnos-
tor systems in individuals with diabetes [1]. tic criteria used.
Cardiovascular autonomic neuropathy (CAN) is In earlier cohorts of patients, particularly
the most studied form of the diabetic autonomic those with long-standing type 1 diabetes (T1D),
neuropathies, and it is a common and deadly potential candidates for pancreas transplantation,
complication of diabetes mellitus, which is fre- abnormalities in cardiovascular reflex tests
quently overlooked in clinical practice due to its (CARTs) suggestive of CAN, were detected in up
characteristic subtle presentation earlier in dis- to 90% of patients [2]. In contrast, prevalence
ease. Yet, timely detection of CAN may help rates are very low in patients with newly diag-
implementation of tailored interventions to pre- nosed T1D, as documented by the primary pre-
vent its progression and mitigate the risk of asso- vention arm of the Diabetes Control and
ciated complications, including cardiovascular Complications Trial (DCCT) cohort, arguably
disease (CVD), cardiac arrhythmias, myocardial the best phenotyped cohort for CAN with CARTs
dysfunction leading to congestive heart failure, as well as with electrocardiograms recordings
and increased mortality [2]. obtained repeatedly over more than 20  years of
follow-up [1–4]. During the observational epide-
miological follow-up of the same cohort, known
as the Epidemiology of Diabetes Interventions
and Complications (EDIC) study, the prevalence
of CAN increased steadily over time to as high as
44% of the cohort over 23 years of mean follow-
L. Ang · R. Pop-Busui (*) ­up [3]. These data are comparable with data
Division of Metabolism, Endocrinology and reported in other observational cohorts with T1D
Diabetes, Department of Internal Medicine, such as the European Insulin-Dependent Diabetes
University of Michigan, Ann Arbor, MI, USA Mellitus Prospective Complications Study
e-mail: rpbusui@umich.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 203
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_12
204 L. Ang and R. Pop-Busui

(EURODIAB IDDM), that reported baseline known T2D duration of 4  years, a mean age of
prevalence rates of autonomic dysfunction, ~57  years, mean HbA1c 8.0% (~64  mmol/mol)
defined as an abnormality in one or both of and diverse ethnic representation presented with
CARTs used, of 36% [5], the STENO T1D study CAN at baseline [11] as diagnosed with the same
that reported similar prevalence of CAN (36%) HRV indices derived from standard 12-lead ECG
over mean duration of diabetes of 40 years using recordings using the recently published cut-­
≥ two abnormal CARTs [6], or the Pittsburgh points based on population norms from the Multi-­
Epidemiology of Diabetes Complications Study Ethnic Study of Atherosclerosis (MESA) [12].
(EDC) study, that reported a cumulative inci- CAN may also present in the pre-diabetic
dence of symptomatic autonomic neuropathy state, in individuals with impaired glucose toler-
over 25  years of approximately 25% [7]. Most ance, insulin resistance, and/or the metabolic
recently, the large contemporary cohort of T1D syndrome as reported from several cohorts dem-
enrolled in the T1D Exchange Clinic Network onstrating the presence of subclinical CAN with
Registry (T1D Exchange) from years 2010–2017, either HRV studies or smaller cardiac sympa-
reported that autonomic neuropathy that included thetic imaging studies [1, 2, 13–16]. Observations
CAN defined with the Survey of Autonomic in the population-based cross-sectional KORA
Symptoms (SAS), was present in ~17% of the S4 study that included more than 1300 partici-
cohort [8]. pants aged 55–74  years phenotyped for CAN
Similar to T1D, prevalence rates as high as with indices of HRV and QT variability, demon-
60% have been reported in earlier cohorts of strated prevalence rates of CAN as high as 8–11%
patients with long-standing T2D [1, 2]. In more among those with prediabetes as defined by
recent T2D cohorts, however, lower prevalence impaired glucose tolerance and or impaired fast-
rates of 9–25% have been observed. For instance, ing glucose, compared with 11.7% in newly diag-
in Europe, the Diabetic Cardiovascular nosed T2D and 17.5% in those with known T2D
Autonomic Neuropathy Multicenter Study Group [17]. Increased heart rate (HR) and decreased
reported CAN prevalence rates of ~22% in T2D HRV were also reported in patients with meta-
using strict criterion of abnormal findings in bolic syndrome, regardless of fasting glucose lev-
CARTs and indices of heart rate variability els [18]. CAN is also observed in youth as
(HRV), while the observational prospective fol- documented by the SEARCH for Diabetes in
low-­up of the Danish arm of the Anglo-Danish-­ Youth Cohort study that included ~2000 youth
Dutch Study of Intensive Treatment in People with T1D and T2D and reported CAN prevalence
With Screen-Detected Diabetes in Primary Care rates of ~12% in youth with T1D (mean age
(ADDITION-Denmark), reported prevalence 18 years, duration of diabetes 8 years) and 17%
rates of 9% and 15% for manifest CAN, defined in youth with T2D (mean age 22 years, duration
based on CARTs, at the 6-year and 13-year fol- of diabetes 8 years) [19].
low-­up examinations [9]. In the USA, in more There are several risk factors that were shown
than 8000 participants with T2D at high CVD to be strongly associated with CAN in both T1D
risk, with a mean age of ~62  years and mean and T2D, with some differences also present
duration of 10  years enrolled in the Action to among these individuals. Notable and consistent
Control Cardiovascular Risk in Diabetes risk factors for CAN across earlier and more con-
(ACCORD) trial; the prevalence of CAN at study temporary cohorts include older age, markers of
entry defined by indices of HRV derived from higher glycemic exposure (including longer dia-
ECG recordings was ~20% [10]. Most recently, betes duration and poor glucose control as docu-
the Glycemia Reduction Approaches in mented by HbA1c), diabetic kidney disease,
Diabetes—A Comparative Effectiveness hypertension, elevated triglycerides, smoking,
(GRADE) trial reported that ~10% of this large and use of some medications [1–3, 20]. Similar
contemporary cohort of ~5000 individuals with risk factors for CAN were also described in youth
T2D of which 36% were female, with a mean [20]. In the highly phenotyped DCCT/EDIC
Cardiovascular Autonomic Neuropathy 205

cohort, in multivariable models, higher mean ney disease, hypertension, elevated triglycer-
HbA1c over time was one of the strongest risk ides, and smoking.
factors for CAN development [3]. Beyond glyce- • Socio-economic factors, female gender, as
mia, in the same cohort, after adjusting for age well as depression are emerging as risk factors
and HbA1c, hypertension and kidney disease for CAN.
(defined as higher albumin excretion ratio and
lower eGFR) as well as higher triglycerides were
independently associated with CAN [3]. 3 Pathophysiology
Hypertension, dyslipidemia and high triglycer-
ides were identified as risk factors for CAN in The potential pathogenic mechanisms leading to
other cohorts such as the EDC and EURODIAB CAN development are broad and may include
IDDM studies for T1D, and as well as in the complex interactions between indices of glyce-
ADDITION study and in American Indians with mic exposure such as chronic hyperglycemia,
T2D [21–25]. impaired insulin signaling, glucose variability
Furthermore, existing evidence has clearly (GV), and disease duration, in conjunction with
established insulin resistance as a positive predic- several other risk factors, including obesity,
tor of abnormal sympathovagal balance in non- hypertension, dyslipidemia and age-related neu-
diabetic offspring of patients with T2D, which ronal deterioration [2]. In concert, these factors
suggests that insulin resistance may be an early activate critical pathways resulting in oxidative
contributor to autonomic dysfunction prior to the stress, low-grade inflammation, and advanced
onset of diabetes [26, 27]. Recent evidence from glycation with resultant consequences on nerve
several large cohorts reported an overall increased cellular metabolism, bioenergetics, and perfusion
CVD risk in women with diabetes, and some [2, 31–34] (Fig. 1).
smaller studies suggest that female sex may also Generally, uncontrolled blood glucose, as
be a risk factor for CAN that confers a higher ini- indicated by HbA1c levels, is thought of as the
tial risk of myocardial injury and dysfunction impetus behind complications development in
[28]. diabetes [35]. Since the results of the DCCT were
More recently, socio-economic factors includ- published [35], intensive glucose control has
ing lower income, and education, as well as been the standard clinical practice in T1D [36].
depression are emerging as risk factors for CAN However, evidence from the DCCT/EDIC study
[8]. The observation of lower prevalence of CAN and other diabetes trials cautions that while main-
in non-White races reported in the GRADE taining near-normal glucose control is important,
cohort adds to an inconsistent body of literature it is not sufficient to prevent all complications nor
on race/ethnicity differences, with population-­ to reverse disease once established [2, 4, 35, 37].
based studies reporting lower, matching, or Furthermore, the observed increase in mortality
higher rates of CAN and other complications in in the intensive glycemic intervention group in
non-White race groups [11, 29, 30]. the ACCORD trial raises the possibility that acute
hypoglycemia and/or wide glucose fluctuations
Key Points were potential mediators of this unexpected out-
come [2, 38]. Historically, observational and
• Cardiovascular autonomic neuropathy is a experimental evidence has implicated hypogly-
prevalent complication in T1D and T2D cemia in the “dead in bed” syndrome, possibly by
including in newly diagnosed T2D. lengthening the QT interval and/or by altering
• High prevalence rates were described in indi- cardiac vagal protection, with relative sympa-
viduals with prediabetes/metabolic syndrome thetic overactivity in T1D and subsequent fatal
as well as in youth. ventricular arrhythmias [2, 39–41]. In addition,
• Notable risk factors for CAN include older individuals with T1D present with disrupted
age, higher glycemic exposure, diabetic kid- compensatory mechanisms in response to hypo-
206 L. Ang and R. Pop-Busui

Hyperglycemia

Hypoglycemia Glucose
Variability

Changes in Oxidative
Metabolic flux Stress

Reactive Inflammation
Nitrogen
Species

Mitochondrial Myeloperoxidase
Pathway; Pathway; Tyrosyl
Superoxide Radical
Hydrogen Hypochlorus
Peroxide Acid

glucoxidation; Endothelial Cell


Polysaturated Fatty Acid; Dysfunction;
Advanced Glycation End Cardivascular Autonomic Superoxide
Products Neuropathy

Cardiac Myocardial
CVD Arrhythmia Heart Failure
Dysfunction

Morbidity Mortality

Fig. 1  The potential complex pathogenic mechanisms of (CVD), cardiac arrhythmia, myocardial dysfunction, and
cardiovascular autonomic neuropathy (CAN) in diabetes. heart failure with subsequent increased morbidity and
CAN may increase the risk of cardiovascular disease mortality

glycemia, such as lack of insulin secretion sup- 49, 50]. Our group has reported that increased
pression, impaired counterregulatory hormones glucose variability, particularly with predomi-
and lack of subjective awareness of hypoglyce- nance of hypoglycemic stress, correlates with
mia, all increasing the risk of cardiac arrhythmia reduced HRV, regardless of HbA1c, in T1D
and sudden cardiac death [2, 42–44]. patients [51], which lends support to a potential
Emerging evidence from animal and human role of high glucose variability and hypoglyce-
studies reported that glucose variability may have mia in development of CAN. In concert with our
more pronounced effects on stimulating several findings, Rao et al. reported a significant decrease
critical pathways involved in the risk of chronic in baroreflex sensitivity, maximum R-R interval
complications including CAN such as: genera- and maximal range of the R-R interval response
tion of reactive oxygen species, endothelial during hypoglycemia induced by a hypoglycemic-­
­dysfunction, and vascular injury [2, 45–48] than hyperinsulinemic clamp (2.8 mmol/L) compared
does chronic hyperglycemia. with baseline euglycemia, illustrating decreased
Moreover, recent human data have suggested parasympathetic influence and disruption in car-
that acute changes in blood glucose levels may be diovascular homeostasis in the setting of hypo-
tied to development of chronic diabetes compli- glycemia [2, 52]. Increased glucose variability
cations such as CAN, independent of HbA1c [2, has been associated with disruptions in sympa-
Cardiovascular Autonomic Neuropathy 207

thovagal balance in T2D, including in well con- diabetic nerve inflammatory response. NF-κB is
trolled patients with T2D without overt autonomic activated by hyperglycemia, oxidative stress, and
neuropathy; therefore, it may be an early marker inflammatory cytokines, thereby playing an
of and contributor to progression of autonomic important role in regulating other inflammatory
dysfunction [2, 53]. In contrast, in the DCCT/ genes and their associated actions, all potentially
EDIC cohort no effect of glucose variability was leading to subsequent deficits in peripheral and
observed on risk of CAN after adjusting for mean autonomic nerve fibers [2, 56, 58–60]. In humans,
blood glucose and other risk factors [2, 54]. our group and others reported significantly higher
However, the limited number of glucose readings markers of inflammation, including TNFα, in
available as part of the seven-point glucose pro- patients with T1D and CAN compared with T1D
files could not accurately characterize the full patients without CAN [2, 31, 33].
spectrum of glucose variability, which is now There is emerging evidence that alterations in
possible with the use of continuous glucose mon- levels of intermediate metabolites of the glyco-
itoring systems (CGMs) [2, 54]. lytic pathway and tricarboxylic acid (TCA) cycle
Several other pathways and mechanisms have are associated with diabetes complications. Our
been documented in the risk for CAN. Oxidative group has recently reported the specific patterns
stress is an important player in the diabetic state, of TCA metabolites and amino acids associated
and contributes to progressive deterioration and with both baseline and changes in measures of
subsequent complications, including CAN, CAN as well as its progression over 3  years of
through several pathways: (1) the glycoxidation follow-up in a cohort with T1D carefully pheno-
and polysaturated fatty acid pathways with for- typed for CAN using CARTs, indices of HRV
mation of advanced glycosylation end-products and cardiac sympathetic imaging with 11C-HED
(AEGs) and reactive intermediates; (2) the reac- PET [2, 61]. There was a significant association
tive nitrogen pathway generating reactive nitro- between higher fumarate and lower asparagine
gen species; (3) the myeloperoxidase pathway and glutamine levels at baseline and measures of
with production of tyrosyl radical hypochlorous CAN. In addition, glutamine and ornithine levels
acid; (4) the mitochondrial pathway with forma- at baseline correlated with CAN progression
tion of superoxide hydrogen peroxide; and (5) after adjusting for diabetes duration and meta-
endothelial cell dysfunction generating excess bolic parameters [2, 61]. Similarly, Ziegler et al.
superoxide [2, 55–57]. Systemic oxidative stress analyzed 127 plasma lipid metabolites using MS
is increased in T1D participants with CAN, and in participants from the German Diabetes Study
cardiac sympathetic tone, as measured by 11C-­ baseline cohort recently diagnosed with either
HED PET, is altered early in the course of T1D T1D or T2D and found that higher plasma levels
[2, 33]. of specific lipid metabolites were closely linked
In addition, both experimental and clinical to cardiac autonomic dysfunction in recent-onset
data have implicated low-grade inflammation as T2D suggesting a role for perturbed lipid metab-
a key mediator in the development of CAN [2, olism in the early development of CAN in T2D
31, 33, 58–60]. In an experimental model of [62]. These most recent reports open the door for
streptozotocin-induced diabetes, Wang et  al. potential therapeutic interventions targeting these
reported that ischemic reperfusion injury (IR) changes to help prevent the development and pro-
resulted in overexpression of the redox-sensitive gression of CAN in individuals with diabetes.
transcription factor, nuclear factor kappa B (NF-­ In summary, the specific mechanisms contrib-
κB), in the sciatic endothelial cell and Schwann uting to diabetic CAN in any given patient remain
cell [2, 60]. This was associated with increased elusive, and results from several clinical trials
expression of intercellular adhesion molecule-1 evaluating the benefit of potential therapeutic
(ICAM-1) and monocyte macrophage infiltration agents targeting proposed pathogenic pathways
in diabetic versus control nerve, lending support in established CAN were disappointing. However,
to NF-κB activation as a key component in the with the emerging progress in diabetes technolo-
208 L. Ang and R. Pop-Busui

gies, as well as big data analyses and integration is an independent predictor of both cardiovascu-
of the genotype/phenotype interactions with mul- lar and all-cause mortality [2, 66, 67]. In a large
tiomics approaches, there is promise for identifi- cardiovascular outcome trial involving ~31,000
cation of specific degrees of susceptibility in an participants with stable cardiovascular disease
individual patient’s risk response [2]. with or without diabetes followed for an average
of 5  years, there was a positive correlation
between HR and both cardiovascular and all-­
4 Clinical Impact cause mortality [2, 68].
and Implications Moreover, the presence of CAN, as docu-
mented by deficits in sympathetic innervation
CAN has a broad spectrum of serious clinical with either iodine-123-labeled metaiodobenzyl-
implications. CAN has emerged as an indepen- guanidine (I-123 MIBG) scintigraphy or
dent predictor for both all-cause and CVD mor- 11C-meta-hydroxyephedrine positron emission
tality [2, 32, 63, 64]. In T1D, earlier studies from tomography (11C-HED PET), was associated
the EURODIAB IDDM cohort showed that with significantly higher mortality rates in
CAN, defined by a lower R–R ratio and postural patients with heart failure (HF) [2, 69, 70]. The
hypotension, was a strong risk marker for mortal- AdreView Myocardial Imaging for Risk
ity over 7  years of follow-up [2, 64]. Similar Evaluation in Heart Failure (ADMIRE-HF)
trends are described for prediabetes and T2D. For study described a positive association between
instance, a sub-study of a glucose tolerance-­ abnormal heart-to-mediastinum ratio (H/M) of
stratified sample of 605 participants from the less than 1.6 on I-123 MIBG scan, used as a
larger Hoorn Study population, which consisted measure of relative myocardial sympathetic
of 2484 individuals aged 50–75 years randomly nerve activity at baseline, and HF progression,
selected from the population register of the Dutch cardiac death, arrhythmic event, and all-cause
town of Hoorn, reported that CAN defined on one mortality in 960 patients with New  York Heart
CART and several HRV indices was associated Association functional class II/III HF over
with a twofold increase in the 9-year mortality 2  years of follow-up [2, 70]. Similarly, the
risk in T2D [2]. In the longitudinal, population-­ Prediction of Arrhythmic Events with Positron
based Monitoring of Trends and Determinants in Emission Tomography (PAREPET) study dem-
Cardiovascular Disease (MONICA)/Cooperative onstrated that baseline deficits in sympathetic
Health Research in the Region of Augsburg innervation on 11C-HED PET were a much stron-
(KORA) cohort study, reduced HRV and pro- ger predictor of sudden cardiac arrest in a cohort
longed corrected QT interval (QTc), both reflec- of patients with ischemic cardiomyopathy (50%
tive of CAN, were associated with excess of whom had diabetes) than either deficits in
mortality in a sample of the diabetic elderly gen- myocardial blood flow or infarct size during
eral population after 9 years of follow-up [2, 65]. 4  years of follow-up [2, 69]. Recent evidence
The conclusive evidence was obtained in greater demonstrated associations between cardiac auto-
than 10,000 participants with T2D in the Action nomic dysfunction, assessed by HRV indices,
to Control Cardiovascular Risk in Diabetes CARTs and particularly cardiac I-123 MIBG
(ACCORD) trial, which reported that the pres- imaging as a marker of cardiac sympathetic
ence of CAN at baseline, defined based on indi- integrity loss, and derangements in myocardial
ces of HRV extracted from 12 leads ECG blood flow regulation in T1D [2, 71]. Therefore,
recordings was an independent predictor of in addition to cardiovagal impairment, cardiac
increased mortality during follow-up after adjust- adrenergic denervation has emerged as a poten-
ing for all other traditional CVD risk factors, and tial risk factor in the development of CAN,
multiple medications [2, 63]. In addition, multi- although the current evidence on its actual role is
ple prospective cohort studies found that an ele- notably less robust and further confirmatory
vated resting HR, a surrogate indicator of CAN, studies are needed [2].
Cardiovascular Autonomic Neuropathy 209

The presence of CAN was also shown to pre- ratio, indices of left ventricular remodeling [2,
dict the development of CVD. The DCCT/EDIC 73]. The prevailing sympathetic activation and
study recently reported that T1D participants imbalance associated with CAN may lead to crit-
diagnosed with CAN at DCCT closeout were at ical changes in myocardial substrate usage result-
significantly higher long-term risk for CVD ing in regional ventricular wall motion
events during follow-up, independent of risk fac- abnormalities and dysfunction [2].
tors, such as age, duration, and BP; although, this Several studies also demonstrated that CAN,
was not independent of glycemic control over defined based on CARTs and QT interval, is a
time as documented by the time-weighted HbA1c risk marker for ischemic stroke [81, 82], with
[1, 2]. In the analysis of ~1100 patients with T2D some studies reporting that CAN was associated
with no symptoms of CVD enrolled in the pro- with up to a twofold increase in stroke risk [2, 34,
spective randomized controlled trial Detection of 74].
Ischemia in Asymptomatic Diabetics (DIAD), Cardiovascular autonomic neuropathy may also
the presence of cardiovascular autonomic dys- contribute to the development of diabetic nephrop-
function, defined by being in the lowest quartile athy in patients with diabetes, with one possible
of HR changes during the lying-to-standing test, mechanism including sympathetic overactivity
was strongly associated with silent myocardial leading to constriction of renal pre-­glomerular ves-
ischemia and subsequent cardiovascular events sels, proteinuria, and further deterioration in renal
[2, 72]. function [2, 83, 84]. More recently, Orlov et  al.
Another important cause of morbidity in CAN reported a strong association between CAN and
is its potential role in the development of myocar- early progressive decline in renal function in
dial dysfunction [32, 73, 74], diabetic cardiomy- patients with T1D [85]. Similar findings have been
opathy [75], and heart failure, which has emerged demonstrated in T2D.  In a cohort study of 204
as a hidden and grossly underestimated compli- patients with T2D, Tahrani et al. found that patients
cation of diabetes [2]. This has been observed in with CAN, identified using HRV testing, had a
both animal and human studies [2, 33, 73, 76, lower mean estimated glomerular filtration rate
77]. In humans, a pilot study in T1D reported that (eGFR) and a higher prevalence rate of albumin-
participants with subclinical microangiopathy uria and chronic kidney disease (CKD) at baseline
demonstrated extensive deficits in left ventricular compared to those without CAN [2, 86].
sympathetic tone, as measured by 11C-HED PET, Furthermore, they established CAN as an indepen-
leading to impaired myocardial blood flow regu- dent predictor of eGFR decline over a subsequent
lation and left ventricular diastolic function [33]. follow-up period of 2.5  years. More recent evi-
Similarly, in a cross-sectional study performed in dence from our group also indicated that CAN is
110 T2D participants with and without CAN, as associated with lesions of diabetic nephropathy as
assessed by CARTs, HRV, and I-123 MIBG scan, documented by kidney biopsy in PIMA Indians
participants with CAN demonstrated signifi- with T2D [2, 87]. In the Atherosclerosis Risk in
cantly reduced diastolic function on resting and Communities (ARIC) study, a prospective observa-
exercise echocardiography [2, 78]. Early stages tional cohort of 13,241 patients followed for a
of diabetic cardiomyopathy in both T1D and T2D median of 16 years, the investigators reported that
are frequently described as deficits in left ventri- increasing resting HR and decreasing HRV corre-
cle relaxation and passive filling, which has been lated with risk of end-stage renal disease (ESRD)
supported by cohort and population-based stud- onset and CKD-­related hospitalization, indepen-
ies [2, 75, 79, 80]. The role of CAN in the devel- dent of diabetes, although notably diabetes was
opment of heart failure is further supported by present in 62% of those who developed ESRD [2,
data obtained from more than 900 participants 88]. The authors also observed a pattern of abnor-
with T1D in the DCCT/EDIC study among mal HRV preceding onset of clinical renal failure
whom the presence of CAN was associated with by many years, speculating the potential role auto-
higher left ventricular mass and mass-to-volume nomic imbalance may play in mediating renal
210 L. Ang and R. Pop-Busui

damage [2]. Another indicator of autonomic imbal- female sexual dysfunction and urinary incontinence
ance is impaired diurnal BP variation, which has among those with worse measure of CAN [101].
been associated with chronic kidney disease devel- These findings suggest that the measures of CAN
opment and progression [2, 89–91]. Abnormalities may be useful for predicting increased risk of
in the physiologic decrease in nocturnal BP female sexual dysfunction and urinary incontinence
­compared to daytime BP, termed non-dipping and development in women withT1D [2, 101]. The
reverse dipping, have displayed predictive potential DCCT/EDIC study also assessed erectile dysfunc-
for progression of nephropathy in T2D, particu- tion via the International Index of Erectile Function
larly ESRD and dialysis [2, 92–95]. Finally, in (IIEF) and lower urinary tract symptoms via the
hemodialysis patients with diabetes who presented American Urological Association Symptom Index
with both CAN and left ventricular hypertrophy (AUASI) in men with T1D [102]. Those with erec-
(LVH), the associated mortality was increased fur- tile dysfunction and/or lower urinary tract symp-
ther compared to when CAN was present alone toms had lower R-R variation and Valsalva ratios
without LVH [2, 96]. than those without supporting that CAN may be a
There is considerable evidence demonstrating risk marker for the onset of urologic complications
that diabetes has early, detrimental effects on reti- in men with long-standing T1D [2, 102].
nal nerve structure and function [2, 97]. Stem et al.
evaluated early neuroretinal changes in patients Key Points
with T1D and reported that T1D patients with no
to moderate diabetic retinopathy displayed • CAN is an independent risk factor for cardio-
changes in inner retina anatomy and physiology, vascular mortality, arrhythmia, silent isch-
as studied through extensive ophthalmic assess- emia, and major cardiovascular events.
ment and optical coherence tomography imaging, • Emerging evidence demonstrates that CAN is
demonstrating the role of retinal autonomic neu- an important risk factor for the development
ropathy in patients with T1D [2, 98, 99]. CAN is of myocardial dysfunction and heart failure in
also associated with increased risk of early prolif- both T1D and T2D that is emerging as an
erative diabetic retinopathy (PDR) in individuals underdiagnosed and important health problem
with diabetes [2, 100]. Krolewski et al. evaluated in diabetes.
the association between autonomic dysfunction, as • CAN is a predictor of the development of dia-
assessed by R-R variation during deep breathing betic kidney disease and may be a risk marker
and BP variation during position changes, and for the onset of urologic complications in both
presence of PDR in insulin-­ dependent diabetes men and women with T1D.
mellitus [100]. They reported that patients with
one abnormal test for CAN (mild CAN) and two
or more abnormal tests (significant CAN) had a 5 Diagnostic
35-fold and 77-fold higher risk of PDR, respec-
tively, compared to those without CAN [2]. Given the above, a timely diagnosis of CAN has
Moreover, there is emerging evidence support- important clinical consequences.
ing a positive association between CAN and devel-
opment of erectile dysfunction (ED) and female
sexual dysfunction in individuals with diabetes [2, 5.1 Screening and Diagnosis
101, 102]. The DCCT/EDIC research group studied in Clinical Care
the association between measures of CAN, as
assessed by CARTs, and female sexual dysfunction It could be quite challenging to detect CAN in its
and urinary incontinence, as evaluated by the early stages in routine clinical care, as patients
Female Sexual Function Index (FSFI-R) and may be completely asymptomatic (subclinical
Sandvik Severity Index, respectively, in 580 women CAN) and may only present with decreased HRV
with T1D reporting that greater odds of developing [1, 2].
Cardiovascular Autonomic Neuropathy 211

5.1.1 Symptoms of CAN symptoms for CAN is quite low requiring an


The most common symptoms of CAN occur upon appropriate differential [1, 2] (Table 1). In addition,
standing and include lightheadedness, weakness, these symptoms occur late in the disease course [1,
palpitations, faintness, and syncope [1, 2] (Table 1). 2]. Providers may also consider screening patients
Thus, targeted questions to unveil the above symp- at risk for developing CAN for hypoglycemia
toms should be included while a medical history is unawareness and vice-versa, as this may be associ-
taken in the office, although the specificity of these ated with CAN [1, 103].

Table 1  Diagnostic algorithm for CAN (Adapted from Pop-Busui et  al, Diabetes Care 2017;40:136–154 with the
permission)?
Symptoms Signs/diagnostic tests Differential work-up
Resting Palpitations could Clinical exam: Resting HR  •  Anemia
Tachycardia be asymptomatic >100 bpm  •  Hyperthyroidism
 •  Fever
 •  CVD (atrial fibrillation, flutter,
other)
 •  Dehydration
 •  Adrenal insufficiency
 •  Medications
  – Sympathomimetic agents
(asthma)
  – Over-the-counter cold
agents containing ephedrine or
pseudoephedrine
  – Dietary supplements (e.g.,
ephedra alkaloids)
 •  Smoking, alcohol, caffeine
 •  Recreational drugs (cocaine,
amphetamines, methamphetamine,
mephedrone)
Orthostatic Light headedness Clinical Exam: A reduction of  •  Adrenal insufficiency
Hypotension Weakness >20 mmHg in the systolic BP or  •  Intravascular volume depletion
Faintness >10 mmHg in diastolic BP within   – Blood loss/Acute anemia
Visual impairment 3 min of standing or upright tilt   – Dehydration
Syncope table testing  •  Pregnancy/postpartum
 •  CVD
   –  Arrhythmias
  – Heart failure
  – Myocarditis
  – Pericarditis
   –  Valvular heart disease
 •  Alcohol
 •  Medications
   –  Antiadrenergics
   –  Antianginals
   –  Antiarrhythmics
   –  Anticholinergics
  – Diuretics
   –  ACEi/ARBs
  – Narcotics
  – Neuroleptics
  – Sedatives
Abnormal BP Non-dipping (lack 24-h BP monitoring
regulation of nocturnal BP
drop)
212 L. Ang and R. Pop-Busui

5.1.2 Clinical Signs of CAN tions may present with symptoms and/or signs
The physiologic HRV is a product of both sympa- mimicking CAN, as illustrated in Table 1 [1, 2].
thetic and parasympathetic activity [1, 2]. Patients
with subclinical CAN present with a decrease in Key Points
HRV, usually with deep breathing or change in
posture, considered the earliest clinical indicator • All patients should be assessed for CAN start-
of CAN [1, 2]. Although HRV testing is largely ing 5 years after diagnosis or in the presence
confined to the research setting, it may be done in of other forms of diabetic neuropathy and/or
the office by either (1) taking an ECG recording other diabetic complications with symptoms
as a patient begins to rise from a seated position or and signs.
(2) taking an ECG recording during 1–2 min of • Tests excluding other comorbidities or drug
deep breathing with calculation of HRV [1]. effects/interactions that could mimic CAN
However, even these relatively simple methods should be performed.
could be challenging in some clinical settings, • Screening for CAN should also be considered
highlighting the need for easily accessible diag- in patients with hypoglycemia unawareness
nostic tools and/or biomarkers for screening and and high glucose variability, prior to insulin
diagnosis of CAN for general clinical care. dose adjustments and/or perioperatively.
As CAN progresses, individuals may present
with resting tachycardia with fixed heart rate
(>100 bpm), exercise intolerance, non-dipping and 5.2 Diagnosis for Clinical
reverse dipping, and in more advanced cases with Research
orthostatic hypotension (a fall in systolic or dia-
stolic blood pressure by >20 mmHg or >10 mmHg, Several tests are available to diagnose CAN
respectively, upon standing without an appropriate including: CARTs, ideally under paced breath-
increase in heart rate) [1, 104]. Orthostatic hypo- ing; HRV studies that derive indices in time and
tension is usually easy to document in the office by frequency domain using various algorithms; 24-h
measuring the blood pressure (BP) supine and BP profiles; baroreflex sensitivity testing; and
after standing, keeping also in mind that in most cardiac sympathetic imaging [1, 2].
cases of CAN, there is no compensatory increase CARTs remain the gold standard for autonomic
in the heart rate, despite hypotension [1]. To docu- testing in both the clinical and research settings [1,
ment non-dipping or reverse dipping, the ambula- 2, 104] and evaluate changes in the heart rate and
tory BP monitoring (ABPM), a standard tool in the BP during several physiological maneuvers such as:
current hypertension management, allows the (1) deep breathing; (2) standing; and (3) a Valsalva
assessment of the diurnal BP patterns that is maneuver [1, 2]. Several factors may influence the
mainly regulated by sleep-awake changes in the results of CARTs, such as intake of caffeine, tobacco
autonomic cardiovascular function [1, 104]. products, food, and some medications like beta
However, it is important to note that none of these blockers. Therefore, these tests should be performed
signs are specific for diabetes related CAN. by uniformly trained personalized personnel and
CAN diagnosis includes documentation of using standardized protocols to minimize variabil-
symptoms and signs of CAN (Table  1). In a ity and bias. In addition, given the very large num-
symptomatic patient with a history of poor glu- ber of recording devices and particularly software
cose control presenting with resting tachycardia ­operating with a “black-box” approach (output of
or postural hypotension, clinicians may not need data without reporting the algorithm that calculated
to perform additional CAN tests given costs and the result) have emerged in recent years, it is neces-
burden after excluding other potential causes [2]. sary to carefully select the most transparent and
Importantly, the diagnosis of CAN requires an verifiable technologies to use for reliable and repro-
extensive differential as many other comorbidi- ducible data. In general, it is currently accepted that
ties, such as cardiovascular disease, adrenal insuf- one abnormal test may indicate early or subclinical
ficiency, thyroid dysfunction, acute blood loss and CAN, while two or more abnormal tests are recom-
anemia, dehydration, and drug effects/interac- mended for a definite diagnosis of CAN [104].
Cardiovascular Autonomic Neuropathy 213

Given that age is one of the most important factors 6 Treatment


affecting HR, age-­related normative are needed for
correct classification settings [1, 2, 104]. As it is the case with diabetic peripheral neuropa-
Indices of HRV are alternative CAN outcome thy, to date no disease modifying agents are
measures that have been used extensively. The available for CAN treatment, likely due to the
HRV can be evaluated either as time domain very complex pathogenic pathways contributing
measures such as: the difference between the lon- to development of CAN [1, 2]. Thus, the manage-
gest and shortest RR interval, the standard devia- ment of CAN includes consideration of glucose
tion of 5-min average of normal RR intervals control and modification of other risk factors.
(SDNN), and the root-mean square of the differ-
ence of successive R–R intervals (rMSSD), or as
frequency domain measures obtained by spectral 6.1 Prevention
analysis or other mathematical algorithms such
as: low, very low and high frequency power [1, 2, In individuals with T1D, the conclusive data
104]. Although traditionally these indices were obtained by the DCCT and later during EDIC study,
initially derived from longer ECG recordings, as well as evidence from several smaller trials
HRV-derived indices from standard 10-s 12-lead strongly support that tight control of blood glucose
ECG recordings have emerged as more feasible implemented as early as possible in the disease
alternative measures of CAN in population stud- course is very effective for preventing CAN and
ies, and have been shown to independently pre- slowing its progression in patients with T1D [1–4,
dict CVD morbidity and mortality in several 106, 107]. For instance, during over an average of
large cohorts of people with and without diabetes 6.5 years DCCT follow-up, intensive glucose con-
[11, 12, 63, 65] and to define prevalence rates. trol, with lower hemoglobin A1c levels, reduced the
Until recently, validation studies evaluating the risk of CAN by 45%, a benefit that persisted during
sensitivity and specificity of these ECG-based EDIC with a 30% reduction in incident CAN over
HRV indices compared to the gold standard an additional 14  years of follow-up [3, 4, 107].
CARTs assessment have not been available. Similarly, the EURODIAB IDDM Complications
However, rigorous assessment of the sensitivity, Study, found a significant correlation between
specificity, and probability of correct classifica- development of CAN and glycemic control [5].
tion of CAN using indices of HRV derived from The evidence for patients with T2D is less
short ECG recordings in predicting CAN com- conclusive, as in several large cohorts intensive
pared to the more laborious method using the control of blood glucose had no effect on the risk
gold standard CARTs were performed in the of CAN [108–110] although one could argue that
DCCT/EDIC cohort that was phenotyped con- these trials may have not included the most reli-
comitantly with both standardized CARTs and able CAN outcomes, and most patients with T2D
digitized ECGs. The indices of HRV had modest enrolled had multiple associated comorbidities
predictive performance comparable with other and other risk factors, such as hypertension,
instruments that have been used successfully to hyperlipidemia, obesity, and require
phenotype diabetic peripheral neuropathy in polypharmacy.
large and diverse cohorts of T1D and T2D [105]. In contrast, glucose control as part of a multi-
Cardiac sympathetic imaging using I-123 factorial intervention that targeted besides hyper-
MIBG scintigraphy or 11C-HED PET, 24-h ambu- glycemia, hypertension, dyslipidemia, and
latory blood pressure profiles, baroreflex sensi- lifestyle, demonstrated a 63% reduction in the
tivity testing to assess cardiac vagal and rate of progression to CAN in a small T2D cohort
sympathetic baroreflex function, and microneu- participating in the STENO-2 trial [111]. Most
rography, while valid and useful in some specific recently, analyses from the ACCORD trial,
research scenarios, require sophisticated infra- reported that after adjusting for multiple other
structure, highly trained personnel, and are quite risk factors, intensive glucose treatment signifi-
expensive and time-consuming widely used in cantly reduced CAN risk by 16% compared to
research protocols [1, 2]. standard intervention in a large cohort of more
214 L. Ang and R. Pop-Busui

than 8000 T2D participants in the ACCORD trial (Table 2). Behavioral supportive measures include:
over a mean 5 years follow-up [10]. These data (a) avoiding abrupt changes in body position; (b)
confirmed a beneficial effect of intensive glyce- avoiding actions that elevate intra-­abdominal and
mic therapy particularly in individuals without intra-thoracic pressures; (c) avoiding medications
cardiovascular disease at baseline. A similar ben- that would exacerbate hypotension such as tricyclic
efit on CAN was found for intensive blood pres- antidepressants, phenothiazines and diuretics; (d)
sure control [10], while intensive treatment of raise the head of the bed during sleep; (e) follow a
hyperlipidemia with statins and fenofibrate did schedule of small and frequent meals to minimize
not. The population-based Coronary Artery Risk postprandial hypotension; (f) consider physical
Development in Young Adults (CARDIA) study counter-­pressure maneuvers such as leg crossing
reported benefit of lifestyle interventions in pre- and squatting; (g) hydrate with fluids and salt, if not
vention of CAN progression [112]. In the contraindicated [1, 2]. Pharmacological therapy
Diabetes Prevention Program (DPP), lifestyle includes midodrine and droxidopa, both of which
interventions improved measures of HRV over are FDA approved for the management of ortho-
time in participants with prediabetes [113]. static hypotension and may be considered in
patients who fail non-pharmacological interven-
tions. However, it is recommended to proceed with
6.2 Symptomatic Treatment a very slow titration and use the minimally effec-
tive dose to avoid undesirable side effects. In
Management of orthostatic hypotension involves selected severe cases low dose fludrocortisone may
both behavioral and pharmacological interventions be also an option [1, 2] (Table 2).

Table 2  Treatment for orthostatic hypotension


Simple measures Physical maneuvers
 •  Avoiding sudden changes in body posture to the  •  Leg crossing
head-up position  •  Squatting
 •  Avoiding medications that aggravate hypotension  •  Muscle pumping
 •  Eating small, frequent meals
 •  Avoiding activities that involve straining
 •  Raising bed head
Pharmacological Dose Major adverse effects Notes
treatment Initial Effective
Midodrine 2.5 mg 2.5–10 mg. p.o, 1–4×/  • Supine –  FDA approved
p.o. day hypertension – Activates α-1 receptors
* First dose to be  •  Piloerection on arterioles and veins,
taken before arising  •  Pruritus increases total peripheral
 •  Paresthesias resistance
 •  Urinary retention
Droxidopa 100 mg 100–600 mg p.o 3×/  • Supine – FDA approved for the
p.o 3×/ day hypertension treatment of neurogenic
day To reduce the potential  •  Headache orthostatic hypotension
for supine  •  Dizziness but not specifically for
hypertension, elevate  •  Nausea individuals with
the head of the bed  • Hyperpyrexia and orthostatic hypotension
and give the last dose Confusion due to diabetes
at least 6 h prior to – α/β adrenergic agonist
bedtime – Effectiveness beyond
2 weeks of treatment has
not been established
Fludrocortisone 0.05 mg 0.1–0.2 mg/day, p.o.  • Supine – Not FDA approved for
po at hypertension orthostatic hypotension
bedtime  •  Hypokalemia – Synthetic
 •  Hypomagnesemia mineralocorticoid
 • Congestive heart – May require up to 1- to
failure 2-week to observe effects
 •  Peripheral edema
Cardiovascular Autonomic Neuropathy 215

7 Conclusions 4. Pop-Busui R, Low PA, Waberski BH, Martin CL,


Albers JW, Feldman EL, Sommer C, Cleary PA,
Lachin JM, Herman WH, Group DER.  Effects of
CAN is prevalent chronic complication of diabe- prior intensive insulin therapy on cardiac auto-
tes with a diverse clinical spectrum of symptoms nomic nervous system function in type 1 diabetes
and signs. The prevalence rates remain high even mellitus: the Diabetes Control and Complications
Trial/Epidemiology of Diabetes Interventions and
in contemporary cohorts and under the current Complications study (DCCT/EDIC). Circulation.
standards of care. Prompt recognition of CAN in 2009;119(22):2886–93.
the clinical setting is crucial, as CAN is an inde- 5. Kempler P, Tesfaye S, Chaturvedi N, Stevens LK,
pendent predictor of cardiovascular mortality, Webb DJ, Eaton S, Kerényi Z, Tamás G, Ward
J, Fuller J.  Autonomic neuropathy is associated
cardiac arrhythmia, silent myocardial ischemia, with increased cardiovascular risk factors: the
major cardiovascular events, and myocardial dys- EURODIAB IDDM Complications Study. Diabet
function leading to increased morbidity, substan- Med. 2002;19(11):900–9.
tial economic burden, and poor quality of life in 6. Hansen C, Theilade S, Lajer M, Hansen T, Rossing
P.  Cardiovascular autonomic neuropathy and
patients with diabetes. Despite this knowledge, bone metabolism in type 1 diabetes. Diabet Med.
screening and diagnosis of the disease in its early 2018;35(11):1596–604.
stages remains elusive, and effective manage- 7. Pambianco G, Costacou T, Ellis D, Becker DJ,
ment strategies and interventions to reverse Klein R, Orchard TJ.  The 30-year natural history
of type 1 diabetes complications: the Pittsburgh
development and progression are missing. The Epidemiology of Diabetes Complications Study
progress in diabetes technologies, combined with experience. Diabetes. 2006;55(5):1463–9.
big data analyses and integration of the genotype/ 8. Mizokami-Stout KR, Bailey R, Foster NC, Ang L,
phenotype interactions with multiomics-derived Aleppo G, Levy CJ, Rickels MR, Shah VN, Polsky
S, Katz M. 563-P: autonomic neuropathy in type 1
biomarkers, are likely to unveil specific increased diabetes (T1D): findings from the T1D exchange.
susceptibilities to or protections against Am Diabetes Assoc. 2019;68(Suppl 1):563.
CAN.  This would also lead to identification of 9. Andersen ST, Witte DR, Fleischer J, Andersen H,
modifiable factors and targeted techniques pro- Lauritzen T, Jorgensen ME, Jensen TS, Pop-Busui
R, Charles M. Risk factors for the presence and pro-
viding the framework for future clinical trials to gression of cardiovascular autonomic neuropathy
test novel interventions to either prevent progres- in type 2 diabetes: ADDITION-Denmark. Diabetes
sion or reverse disease in a personalized care Care. 2018;41(12):2586–94.
approach, ultimately reducing the health care 10. Tang Y, Shah H, Bueno Junior CR, Sun X, Mitri J,
Sambataro M, Sambado L, Gerstein HC, Fonseca V,
burden and optimizing patients’ quality of life. Doria A, Pop-Busui R. Intensive risk factor manage-
ment and cardiovascular autonomic neuropathy in
type 2 diabetes: the ACCORD trial. Diabetes Care.
References 2021;44(1):164–73.
11. Mather KJ, Bebu I, Baker C, Cohen RM, Crandall
JP, DeSouza C, Green JB, Kirkman MS, Krause-­
1. Pop-Busui R, Boulton AJM, Feldman EL, Bril
Steinrauf H, Larkin M.  Prevalence of microvas-
V, Freeman R, Malik RA, Sosenko JM, Ziegler
cular and macrovascular disease in the Glycemia
D.  Diabetic neuropathy: a position statement by
Reduction Approaches in Diabetes-A Comparative
the American Diabetes Association. Diabetes Care.
Effectiveness (GRADE) Study cohort. Diabetes Res
2017;40(1):136–54.
Clin Pract. 2020;165:108235.
2. Ang L, Dillon B, Mizokami-Stout K, Pop-Busui
12. O’Neal WT, Chen LY, Nazarian S, Soliman
R.  Cardiovascular autonomic neuropathy: a
EZ. Reference ranges for short-term heart rate vari-
silent killer with long reach. Auton Neurosci.
ability measures in individuals free of cardiovascular
2020;225:102646.
disease: the Multi-Ethnic Study of Atherosclerosis
3. Braffett BH, Gubitosi-Klug RA, Albers JW,
(MESA). J Electrocardiol. 2016;49(5):686–90.
Feldman EL, Martin CL, White NH, Orchard TJ,
13. Diakakis GF, Parthenakis FI, Patrianakos AP,
Lopes-Virella M, Lachin JM, Pop-Busui R.  Risk
Koukouraki SI, Stathaki MI, Karkavitsas NS,
factors for diabetic peripheral neuropathy and car-
Vardas PE.  Myocardial sympathetic innervation in
diovascular autonomic neuropathy in the Diabetes
patients with impaired glucose tolerance: relation-
Control and Complications Trial/Epidemiology of
ship to subclinical inflammation. Cardiovasc Pathol.
Diabetes Interventions and Complications (DCCT/
2008;17(3):172–7.
EDIC) study. Diabetes. 2020;69(5):1000–10.
216 L. Ang and R. Pop-Busui

14. Papanas N, Vinik AI, Ziegler D. Neuropathy in pre- 25. Reynolds EL, Akinci G, Banerjee M, Looker HC,
diabetes: does the clock start ticking early? Nat Rev Patterson A, Nelson RG, Feldman EL, Callaghan
Endocrinol. 2011;7(11):682. BC. The determinants of complication trajectories in
15. Putz Z, Tabák ÁG, Tóth N, Istenes I, Németh N, American Indians with type 2 diabetes. JCI Insight.
Gandhi RA, Hermányi Z, Keresztes K, Jermendy G, 2021;6(10):e146849.
Tesfaye S. Noninvasive evaluation of neural impair- 26. Frontoni S, Bracaglia D, Baroni A, Pellegrini F,
ment in subjects with impaired glucose tolerance. Perna M, Cicconetti E, Ciampittiello G, Menzinger
Diabetes Care. 2009;32(1):181–3. G, Gambardella S.  Early autonomic dysfunc-
16. Wu J-S, Yang Y-C, Lin T-S, Huang Y-H, Chen J-J, Lu tion in glucose-tolerant but insulin-resistant off-
F-H, Wu C-H, Chang C-J. Epidemiological evidence spring of type 2 diabetic patients. Hypertension.
of altered cardiac autonomic function in subjects 2003;41(6):1223–7.
with impaired glucose tolerance but not isolated 27. Laitinen T, Vauhkonen I, Niskanen LK, Hartikainen
impaired fasting glucose. J Clin Endocrinol Metab. J, Länsimies EA, Uusitupa M, Laakso M.  Power
2007;92(10):3885–9. spectral analysis of heart rate variability during
17. Ziegler D, Voss A, Rathmann W, Strom A, Perz S, hyperinsulinemia in nondiabetic offspring of type 2
Roden M, Peters A, Meisinger C.  Increased preva- diabetic patients: evidence for possible early auto-
lence of cardiac autonomic dysfunction at differ- nomic dysfunction in insulin-resistant subjects.
ent degrees of glucose intolerance in the general Diabetes. 1999;48(6):1295–9.
population: the KORA S4 survey. Diabetologia. 28. Duvernoy CS, Raffel DM, Swanson SD, Jaiswal
2015;58(5):1118–28. M, Mueller G, Ibrahim E-S, Pennathur S, Plunkett
18. Stein P, Barzilay J, Domitrovich P, Chaves P, C, Stojanovska J, Brown MB, Pop-Busui R.  Left
Gottdiener J, Heckbert S, Kronmal R. The relation- ventricular metabolism, function, and sympathetic
ship of heart rate and heart rate variability to non-­ innervation in men and women with type 1 diabetes.
diabetic fasting glucose levels and the metabolic J Nucl Cardiol. 2016;23(5):960–9.
syndrome: the Cardiovascular Health Study. Diabet 29. Winkley K, Thomas S, Sivaprasad S, Chamley
Med. 2007;24(8):855–63. M, Stahl D, Ismail K, Amiel S.  The clinical
19. Jaiswal M, Divers J, Urbina EM, Dabelea D, Bell characteristics at diagnosis of type 2 diabetes
­
RA, Pettitt DJ, Imperatore G, Pihoker C, Dolan LM, in a multi-­ ethnic population: the South London
Liese AD, Marcovina S, Linder B, Feldman EL, Diabetes cohort (SOUL-D). Diabetologia.
Pop-Busui R, Group SfDiYS. Cardiovascular auto- 2013;56(6):1272–81.
nomic neuropathy in adolescents and young adults 30. Tahrani AA, Altaf QA, Piya MK, Barnett
with type 1 and type 2 diabetes: the SEARCH for AH. Peripheral and autonomic neuropathy in South
Diabetes in Youth Cohort Study. Pediatr Diabetes. Asians and White Caucasians with type 2 diabetes
2018;19(4):680–9. mellitus: possible explanations for epidemiological
20. Dabelea D, Stafford JM, Mayer-Davis EJ, differences. J Diabetes Res. 2017;2017:1273789.
D’Agostino R, Dolan L, Imperatore G, Linder 31. Lieb DC, Parson HK, Mamikunian G, Vinik
B, Lawrence JM, Marcovina SM, Mottl AI.  Cardiac autonomic imbalance in newly diag-
AK.  Association of type 1 diabetes vs type 2 dia- nosed and established diabetes is associated with
betes diagnosed during childhood and adolescence markers of adipose tissue inflammation. Exp
with complications during teenage years and young Diabetes Res. 2012;2012:8.
adulthood. JAMA. 2017;317(8):825–35. 32. Pop-Busui R. What do we know and we do not know
21. Maser RE, Pfeifer MA, Dorman JS, Kuller LH, about cardiovascular autonomic neuropathy in dia-
Becker DJ, Orchard TJ.  Diabetic autonomic betes. J Cardiovasc Transl Res. 2012;5(4):463–78.
neuropathy and cardiovascular risk: Pittsburgh 33. Pop-Busui R, Kirkwood I, Schmid H, Marinescu
Epidemiology of Diabetes Complications Study V, Schroeder J, Larkin D, Yamada E, Raffel DM,
III. JAMA Intern Med. 1990;150(6):1218–22. Stevens MJ. Sympathetic dysfunction in type 1 dia-
22. Stella P, Ellis D, Maser RE, Orchard betes: association with impaired myocardial blood
TJ.  Cardiovascular autonomic neuropathy (expi- flow reserve and diastolic dysfunction. J Am Coll
ration and inspiration ratio) in type 1 diabetes: Cardiol. 2004;44(12):2368–74.
incidence and predictors. J Diabetes Complicat. 34. Vinik AI, Ziegler D.  Diabetic cardiovascular auto-
2000;14(1):1–6. nomic neuropathy. Circulation. 2007;115(3):387–97.
23. Tesfaye S, Chaturvedi N, Eaton SE, Ward JD, 35. Nathan DM, Genuth S, Lachin J, Cleary P, Crofford
Manes C, Ionescu-Tirgoviste C, Witte DR, Fuller O, Davis M, Rand L, Siebert C. The effect of inten-
JH. Vascular risk factors and diabetic neuropathy. N sive treatment of diabetes on the development
Engl J Med. 2005;352(4):341–50. and progression of long-term complications in
24. Andersen ST, Witte DR, Andersen H, Bjerg L, insulin-­dependent diabetes mellitus. N Engl J Med.
Bruun NH, Jørgensen ME, Finnerup NB, Lauritzen 1993;329(14):977–86.
T, Jensen TS, Tankisi H.  Risk-factor trajectories 36. Glycemic targets: standards of medical care in
preceding diabetic polyneuropathy: ADDITION-­ diabetes—2021. Diabetes Care. 2021;44(Suppl.
Denmark. Diabetes Care. 2018;41(9):1955–62. 1):S73–84.
Cardiovascular Autonomic Neuropathy 217

37. Ang L, Jaiswal M, Martin C, Pop-Busui R. Glucose patients with type 1 diabetes. Diabetes Care.
control and diabetic neuropathy: lessons from recent 2014;37(9):2616–21.
large clinical trials. Curr Diab Rep. 2014;14(9):1–15. 52. Rao AD, Bonyhay I, Dankwa J, Baimas-George
38. Gerstein HC, Miller ME, Byington RP, Goff DC Jr, M, Kneen L, Ballatori S, Freeman R, Adler
Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-­ GK. Baroreflex sensitivity impairment during hypo-
Beigi F, Grimm RH Jr, Probstfield JL, Simons-­ glycemia: implications for cardiovascular control.
Morton DG, Friedewald WT.  Effects of intensive Diabetes. 2016;65(1):209–15.
glucose lowering in type 2 diabetes. N Engl J Med. 53. Di Flaviani A, Picconi F, Di Stefano P, Giordani I,
2008;358(24):2545–59. Malandrucco I, Maggio P, Palazzo P, Sgreccia F,
39. Gill GV, Woodward A, Casson IF, Weston Peraldo C, Farina F. Impact of glycemic and blood
PJ.  Cardiac arrhythmia and nocturnal hypoglycae- pressure variability on surrogate measures of car-
mia in type 1 diabetes—the ‘dead in bed’ syndrome diovascular outcomes in type 2 diabetic patients.
revisited. Diabetologia. 2009;52(1):42–5. Diabetes Care. 2011;34(7):1605–9.
40. Tu E, Twigg SM, Semsarian C. Sudden death in type 54. Lachin JM, Bebu I, Bergenstal RM, Pop-Busui
1 diabetes: the mystery of the ‘dead in bed’ syn- R, Service FJ, Zinman B, Nathan DM, Group
drome. Int J Cardiol. 2010;138(1):91–3. DER.  Association of glycemic variability in type
41. Weston PJ, Gill GV.  Is undetected autonomic dys- 1 diabetes with progression of microvascular out-
function responsible for sudden death in type 1 comes in the diabetes control and complications
diabetes mellitus? The ‘dead in bed’ syndrome revis- trial. Diabetes Care. 2017;40(6):777–83.
ited. Diabet Med. 1999;16(8):626–31. 55. Hoeldtke RD, Bryner KD, VanDyke K.  Oxidative
42. Cryer PE, Davis SN, Shamoon H. Hypoglycemia in stress and autonomic nerve function in early type 1
diabetes. Diabetes Care. 2003;26(6):1902–12. diabetes. Clin Auton Res. 2011;21(1):19–28.
43. Desouza CV, Bolli GB, Fonseca V.  Hypoglycemia, 56. Kellogg AP, Wiggin TD, Larkin DD, Hayes JM,
diabetes, and cardiovascular events. Diabetes Care. Stevens MJ, Pop-Busui R.  Protective effects of
2010;33(6):1389–94. cyclooxygenase-2 gene inactivation against periph-
44. Limberg J, Farni K, Taylor J, Dube S, Basu A, Basu eral nerve dysfunction and intraepidermal nerve
R, Wehrwein E, Joyner M. Autonomic control dur- fiber loss in experimental diabetes. Diabetes.
ing acute hypoglycemia in type 1 diabetes mellitus. 2007;56(12):2997–3005.
Clin Auton Res. 2014;24(6):275–83. 57. Pennathur S, Heinecke JW.  Mechanisms for oxi-
45. Ceriello A, Esposito K, Piconi L, Ihnat MA, Thorpe dative stress in diabetic cardiovascular disease.
JE, Testa R, Boemi M, Giugliano D.  Oscillating Antioxid Redox Signal. 2007;9(7):955–69.
glucose is more deleterious to endothelial func- 58. Cameron NE, Cotter MA.  Pro-inflammatory
tion and oxidative stress than mean glucose in mechanisms in diabetic neuropathy: focus on the
normal and type 2 diabetic patients. Diabetes. nuclear factor kappa B pathway. Curr Drug Targets.
2008;57(5):1349–54. 2008;9(1):60–7.
46. Horváth E, Benkő R, Kiss L, Muranyi M, Pek T, 59. Pop-Busui R, Ang L, Holmes C, Gallagher K,
Fekete K, Barany T, Somlai A, Csordas A, Szabo Feldman EL.  Inflammation as a therapeutic tar-
C.  Rapid ‘glycaemic swings’ induce nitrosative get for diabetic neuropathies. Curr Diab Rep.
stress, activate poly (ADP-ribose) polymerase and 2016;16(3):29.
impair endothelial function in a rat model of diabetes 60. Wang Y, Schmeichel AM, Iida H, Schmelzer JD,
mellitus. Diabetologia. 2009;52(5):952–61. Low PA. Enhanced inflammatory response via acti-
47. Monnier L, Mas E, Ginet C, et al. Activation of oxi- vation of NF-κB in acute experimental diabetic neu-
dative stress by acute glucose fluctuations compared ropathy subjected to ischemia–reperfusion injury. J
with sustained chronic hyperglycemia in patients Neurol Sci. 2006;247(1):47–52.
with type 2 diabetes. JAMA. 2006;295(14):1681–7. 61. Mathew AV, Jaiswal M, Ang L, Michailidis G,
48. Saisho Y. Glycemic variability and oxidative stress: Pennathur S, Pop-Busui R.  Impaired amino acid
a link between diabetes and cardiovascular disease? and TCA metabolism and cardiovascular autonomic
Int J Mol Sci. 2014;15(10):18381–406. neuropathy progression in type 1 diabetes. Diabetes.
49. Fleischer J.  Diabetic autonomic imbalance and 2019;68(10):2035–44.
glycemic variability. J Diabetes Sci Technol. 62. Ziegler D, Strom A, Straßburger K, Knebel B, Bönhof
2012;6(5):1207–15. GJ, Kotzka J, Szendroedi J, Roden M. Association
50. Fleischer J, Cichosz SL, Hoeyem P, Laugesen E, of cardiac autonomic dysfunction with higher levels
Poulsen PL, Christiansen JS, Tarnow L, Hansen of plasma lipid metabolites in recent-onset type 2
TK. Glycemic variability is associated with reduced diabetes. Diabetologia. 2021;64(2):458–68.
cardiac autonomic modulation in women with type 2 63. Pop-Busui R, Evans GW, Gerstein HC, Fonseca V,
diabetes. Diabetes Care. 2015;38(4):682–8. Fleg JL, Hoogwerf BJ, Genuth S, Grimm RH, Corson
51. Jaiswal M, McKeon K, Comment N, Henderson MA, Prineas R, Group tAS. Effects of cardiac auto-
J, Swanson S, Plunkett C, Nelson P, Pop-Busui nomic dysfunction on mortality risk in the Action to
R.  Association between impaired cardiovascu- Control Cardiovascular Risk in Diabetes (ACCORD)
lar autonomic function and hypoglycemia in trial. Diabetes Care. 2010;33(7):1578–84.
218 L. Ang and R. Pop-Busui

64. Soedamah-Muthu SS, Chaturvedi N, Witte DR, Complications Trial/Epidemiology of Diabetes


Stevens LK, Porta M, Fuller JH.  Relationship Interventions and Complications). J Am Coll
between risk factors and mortality in type 1 diabetic Cardiol. 2013;61(4):447–54.
patients in Europe: the EURODIAB Prospective 74. Spallone V, Ziegler D, Freeman R, Bernardi L,
Complications Study (PCS). Diabetes Care. Frontoni S, Pop-Busui R, Stevens M, Kempler P,
2008;31(7):1360–6. Hilsted J, Tesfaye S, Low P, Valensi P, On behalf
65. Ziegler D, Zentai CP, Perz S, Rathmann W, Haastert of The Toronto Consensus Panel on Diabetic
B, Döring A, Meisinger C.  Prediction of mortal- N.  Cardiovascular autonomic neuropathy in
ity using measures of cardiac autonomic dysfunc- diabetes: clinical impact, assessment, diagno-
tion in the diabetic and nondiabetic population: the sis, and management. Diabetes Metab Res Rev.
MONICA/KORA Augsburg Cohort Study. Diabetes 2011;27(7):639–53.
Care. 2008;31(3):556–61. 75. Fang ZY, Prins JB, Marwick TH.  Diabetic cardio-
66. Ho JE, Bittner V, DeMicco DA, Breazna A, myopathy: evidence, mechanisms, and therapeutic
Deedwania PC, Waters DD. Usefulness of heart rate implications. Endocr Rev. 2004;25(4):543–67.
at rest as a predictor of mortality, hospitalization 76. Mizushige K, Yao L, Noma T, Kiyomoto H, Yu Y,
for heart failure, myocardial infarction, and stroke Hosomi N, Ohmori K, Matsuo H. Alteration in left
in patients with stable coronary heart disease (Data ventricular diastolic filling and accumulation of
from the Treating to New Targets [TNT] trial). Am J myocardial collagen at insulin-resistant prediabetic
Cardiol. 2010;105(7):905–11. stage of a type II diabetic rat model. Circulation.
67. Nauman J, Janszky I, Vatten LJ, Wisløff U. Temporal 2000;101(8):899–907.
changes in resting heart rate and deaths from isch- 77. Semeniuk LM, Kryski AJ, Severson
emic heart disease. JAMA. 2011;306(23):2579–87. DL.  Echocardiographic assessment of cardiac
68. Lonn EM, Rambihar S, Gao P, Custodis FF, Sliwa K, function in diabetic db/db and transgenic db/
Teo KK, Yusuf S, Böhm M. Heart rate is associated db-hGLUT4 mice. Am J Phys Heart Circ Phys.
with increased risk of major cardiovascular events, 2002;283(3):H976–82.
cardiovascular and all-cause death in patients with 78. Sacre JW, Franjic B, Jellis CL, Jenkins C, Coombes
stable chronic cardiovascular disease: an analysis JS, Marwick TH. Association of cardiac autonomic
of ONTARGET/TRANSCEND.  Clin Res Cardiol. neuropathy with subclinical myocardial dysfunc-
2014;103(2):149–59. tion in type 2 diabetes. JACC Cardiovasc Imaging.
69. Fallavollita JA, Heavey BM, Luisi AJ Jr, Michalek 2010;3(12):1207–15.
SM, Baldwa S, Mashtare TL Jr, Hutson AD, Dekemp 79. Astorri E, Fiorina P, Astorri A, Contini GA, Albertini
RA, Haka MS, Sajjad M, Cimato TR, Curtis AB, D, Magnati G, Lanfredini M. Isolated and preclini-
Cain ME, Canty JM Jr. Regional myocardial sym- cal impairment of left ventricular filling in insulin-­
pathetic denervation predicts the risk of sudden car- dependent and non-insulin-dependent diabetic
diac arrest in ischemic cardiomyopathy. J Am Coll patients. Clin Cardiol. 1997;20(6):536–40.
Cardiol. 2014;63(2):141–9. 80. Schannwell CM, Schneppenheim M, Perings S,
70. Jacobson AF, Senior R, Cerqueira MD, Wong Plehn G, Strauer B.  Left ventricular diastolic dys-
ND, Thomas GS, Lopez VA, Agostini D, Weiland function as an early manifestation of diabetic cardio-
F, Chandna H, Narula J.  Myocardial iodine-123 myopathy. Cardiology. 2002;98(1-2):33–9.
meta-iodobenzylguanidine imaging and cardiac 81. Cohen JA, Estacio RO, Lundgren RA, Esler AL,
events in heart failure: results of the prospective Schrier RW. Diabetic autonomic neuropathy is asso-
ADMIRE-HF (AdreView Myocardial Imaging for ciated with an increased incidence of strokes. Auton
Risk Evaluation in Heart Failure) study. J Am Coll Neurosci. 2003;108(1):73–8.
Cardiol. 2010;55(20):2212–21. 82. Ko S, Song K, Park S, Kim S, Cha B, Son H, Moon
71. Zobel EH, Hasbak P, Winther SA, Hansen CS, K, Yoo K, Park Y, Cho J.  Cardiovascular auto-
Fleischer J, von Scholten BJ, Holmvang L, Kjaer A, nomic dysfunction predicts acute ischaemic stroke
Rossing P, Hansen TW. Cardiac autonomic function in patients with type 2 diabetes mellitus: a 7-year
is associated with myocardial flow reserve in type 1 follow-up study. Diabet Med. 2008;25(10):1171–7.
diabetes. Diabetes. 2019;68(6):1277–86. 83. Axelrod S, Lishner M, Oz O, Bernheim J, Ravid
72. Young LH, Frans JT, Chyun DA, Davey JA, Barrett M. Spectral analysis of fluctuations in heart rate: an
EJ, Taillefer R, Heller GV, Iskandrian AE, Wittlin objective evaluation of autonomic nervous control in
SD, Filipchuk N. Cardiac outcomes after screening chronic renal failure. Nephron. 1987;45(3):202–6.
for asymptomatic coronary artery disease in patients 84. Converse RL Jr, Jacobsen TN, Toto RD, Jost CM,
with type 2 diabetes: the DIAD study: a randomized Cosentino F, Fouad-Tarazi F, Victor RG. Sympathetic
controlled trial. JAMA. 2009;301(15):1547–55. overactivity in patients with chronic renal failure. N
73. Pop-Busui R, Cleary PA, Braffett BH, Martin Engl J Med. 1992;327(27):1912–8.
CL, Herman WH, Low PA, Lima JAC, Bluemke 85. Orlov S, Cherney DZ, Pop-Busui R, Lovblom LE,
DA.  Association between cardiovascular auto- Ficociello LH, Smiles AM, Warram JH, Krolewski
nomic neuropathy and left ventricular dysfunc- AS, Perkins BA. Cardiac autonomic neuropathy and
tion: DCCT/EDIC study (Diabetes Control and early progressive renal decline in patients with non-
Cardiovascular Autonomic Neuropathy 219

macroalbuminuric type 1 diabetes. Clin J Am Soc retinal sensory neuropathy in persons with type 1
Nephrol. 2015;10(7):1136–44. diabetes mellitus. Eye (Lond). 2016;30(6):825–32.
86. Tahrani AA, Dubb K, Raymond NT, Begum S, Altaf 99. Antonetti DA, Klein R, Gardner TW. Diabetic reti-
QA, Sadiqi H, Piya MK, Stevens MJ. Cardiac auto- nopathy. N Engl J Med. 2012;366(13):1227–39.
nomic neuropathy predicts renal function decline 100. Krolewski AS, Barzilay J, Warram JH, Martin BC,
in patients with type 2 diabetes: a cohort study. Pfeifer M, Rand LI.  Risk of early-onset prolif-
Diabetologia. 2014;57(6):1249–56. erative retinopathy in IDDM is closely related to
87. Wheelock KM, Jaiswal M, Martin CL, Fufaa GD, cardiovascular autonomic neuropathy. Diabetes.
Weil EJ, Lemley KV, Yee B, Feldman E, Brosius 1992;41(4):430–7.
FC III, Knowler WC.  Cardiovascular autonomic 101. Hotaling JM, Sarma AV, Patel DP, Braffett BH, Cleary
neuropathy associates with nephropathy lesions in PA, Feldman E, Herman WH, Martin CL, Jacobson
American Indians with type 2 diabetes. J Diabetes AM, Wessells H, Pop-Busui R.  Cardiovascular
Complicat. 2016;30(5):873–9. autonomic neuropathy, sexual dysfunction, and uri-
88. Brotman DJ, Bash LD, Qayyum R, Crews D, nary incontinence in women with type 1 diabetes.
Whitsel EA, Astor BC, Coresh J. Heart rate variabil- Diabetes Care. 2016;39(9):1587–93.
ity predicts ESRD and CKD-related hospitalization. 102. Pop-Busui R, Hotaling J, Braffett BH, Cleary PA,
J Am Soc Nephrol. 2010;21(9):1560–70. Dunn RL, Martin CL, Jacobson AM, Wessells H,
89. Davidson MB, Hix JK, Vidt DG, Brotman Sarma AV, Group DER.  Cardiovascular autonomic
DJ.  Association of impaired diurnal blood pres- neuropathy, erectile dysfunction and lower urinary
sure variation with a subsequent decline in tract symptoms in men with type 1 diabetes: findings
glomerular filtration rate. Arch Intern Med. from the DCCT/EDIC. J Urol. 2015;193(6):2045–51.
2006;166(8):846–52. 103. Lin YK, Fisher SJ, Pop-Busui R.  Hypoglycemia
90. Farmer C, Goldsmith D, Quin JD, Dallyn P, Cox J, unawareness and autonomic dysfunction in diabetes:
Kingswood JC, Sharpstone P. Progression of diabetic lessons learned and roles of diabetes technologies. J
nephropathy—is diurnal blood pressure rhythm as Diabetes Investig. 2020;11(6):1388–402.
important as absolute blood pressure level? Nephrol 104. Spallone V, Bellavere F, Scionti L, Maule S,
Dial Transplant. 1998;13(3):635–9. Quadri R, Bax G, Melga P, Viviani GL, Esposito K,
91. Lurbe E, Redon J, Kesani A, Pascual JM, Tacons J, Morganti R, Cortelli P, Diabetic Neuropathy Study
Alvarez V, Batlle D. Increase in nocturnal blood pres- Group of the Italian Society of D. Recommendations
sure and progression to microalbuminuria in type 1 for the use of cardiovascular tests in diagnosing dia-
diabetes. N Engl J Med. 2002;347(11):797–805. betic autonomic neuropathy. Nutr Metab Cardiovasc
92. Knudsen ST, Laugesen E, Hansen K, Bek T, Dis. 2011;21(1):69–78.
Mogensen C, Poulsen P.  Ambulatory pulse pres- 105. Pop-Busui R, Backlund JYC, Bebu I, Braffett BH,
sure, decreased nocturnal blood pressure reduction Lorenzi G, White NH, Lachin JM, Soliman EZ,
and progression of nephropathy in type 2 diabetic Group DER. The utility of using ECG measures of
patients. Diabetologia. 2009;52(4):698–704. heart rate variability as a measure of cardiovascular
93. Nakano S, Ogihara M, Tamura C, Kitazawa M, autonomic neuropathy in type 1 diabetes. J Diabetes
Nishizawa M, Kigoshi T, Uchida K. Reversed circa- Investig. 2022;13(1):125–33.
dian blood pressure rhythm independently predicts 106. Margolis KL, O’Connor PJ, Morgan TM, Buse JB,
endstage renal failure in non–insulin-dependent Cohen RM, Cushman WC, Cutler JA, Evans GW,
diabetes mellitus subjects. J Diabetes Complicat. Gerstein HC, Grimm RH Jr, Lipkin EW, Venkat
1999;13(4):224–31. Narayan KM, Riddle MC Jr, Sood A, Goff DC Jr.
94. Palmas W, Pickering T, Teresi J, Schwartz JE, Eguchi Outcomes of combined cardiovascular risk fac-
K, Field L, Weinstock RS, Shea S. Nocturnal blood tor management strategies in type 2 diabetes:
pressure elevation predicts progression of albumin- the ACCORD randomized trial. Diabetes Care.
uria in elderly people with type 2 diabetes. J Clin 2014;37(6):1721–8.
Hypertens. 2008;10(1):12–20. 107. Martin CL, Albers JW, Pop-Busui R.  Neuropathy
95. Spallone V. Blood pressure variability and autonomic and related findings in the diabetes control and
dysfunction. Curr Diab Rep. 2018;18(12):1–14. complications trial/epidemiology of diabetes inter-
96. Nishimura M, Hashimoto T, Kobayashi H, Fukuda ventions and complications study. Diabetes Care.
T, Okino K, Yamamoto N, Nakamura N, Yoshikawa 2014;37(1):31–8.
T, Takahashi H, Ono T. Association between cardio- 108. Azad N, Emanuele NV, Abraira C, Henderson WG,
vascular autonomic neuropathy and left ventricu- Colwell J, Levin SR, Nuttall FQ, Comstock JP, Sawin
lar hypertrophy in diabetic haemodialysis patients. CT, Silbert C, Rubino FA.  The effects of intensive
Nephrol Dial Transplant. 2004;19(10):2532–8. glycemic control on neuropathy in the VA coopera-
97. Simó R, Hernández C. Neurodegeneration is an early tive study on type II diabetes mellitus (VA CSDM). J
event in diabetic retinopathy: therapeutic implica- Diabetes Complicat. 1999;13(5-6):307–13.
tions. Br J Ophthalmol. 2012;96(10):1285–90. 109. Charles M, Fleischer J, Witte DR, Ejskjaer N, Borch-­
98. Stem MS, Dunbar GE, Jackson GR, Farsiu S, Pop-­ Johnsen K, Lauritzen T, Sandbaek A. Impact of early
Busui R, Gardner TW. Glucose variability and inner detection and treatment of diabetes on the 6-year
220 L. Ang and R. Pop-Busui

prevalence of cardiac autonomic neuropathy in vascular disease in patients with type 2 diabetes. N
people with screen-detected diabetes: ADDITION-­ Engl J Med. 2003;348(5):383–93.
Denmark, a cluster-randomised study. Diabetologia. 112. Carnethon MR, Jacobs DR Jr, Sidney S, Sternfeld
2013;56(1):101–8. B, Gidding SS, Shoushtari C, Liu K.  A longitudi-
110. Duckworth W, Abraira C, Moritz T, Reda D, nal study of physical activity and heart rate recov-
Emanuele N, Reaven PD, Zieve FJ, Marks J, Davis ery: CARDIA, 1987-1993. Med Sci Sports Exerc.
SN, Hayward R, Warren SR, Goldman S, McCarren 2005;37(4):606–12.
M, Vitek ME, Henderson WG, Huang GD. Glucose 113. Carnethon MR, Prineas RJ, Temprosa M, Zhang
control and vascular complications in veterans with ZM, Uwaifo G, Molitch ME. The association among
type 2 diabetes. N Engl J Med. 2009;360(2):129–39. autonomic nervous system function, incident diabe-
111. Gaede P, Vedel P, Larsen N, Jensen GV, Parving HH, tes, and intervention arm in the diabetes prevention
Pedersen O. Multifactorial intervention and cardio- program. Diabetes Care. 2006;29(4):914–9.
Psychosocial Aspects of Diabetic
Neuropathy: From Description
to Interventions

Loretta Vileikyte and Frans Pouwer

1 Introduction matory cytokine system, and a host of other,


interrelated physiological pathways in brain and
The diabetic neuropathies are a collection of body, reviewed elsewhere [5, 6]. This chapter
nerve disorders caused by diabetes that affect dif- focuses exclusively on psychosocial factors asso-
ferent parts of the nervous system and present ciated with DPN.
with diverse clinical manifestations of sensorim- Depression has received the lion’s share of
otor and autonomic dysfunction [1]. Distal sym- attention from clinical researchers. In people
metric polyneuropathy (DPN) is one of the with diabetes, those with co-morbid complica-
commonest varieties and is the focus of this tions are at increased risk to be depressed or
chapter. Among the various clinical presentations develop depression [7–9]. In two longitudinal
of DPN, severe unremitting pain is by far the studies, people with both diabetes and depression
most studied by behavioral scientists. Our chap- had a higher risk to develop DPN [10, 11].
ter, while reflecting on this common tendency, Longitudinal data from nationwide general prac-
also highlights the importance of other clinical tices in Germany showed that individuals with
presentations of DPN.  Postural instability, for diabetes and DPN had higher levels of incident
example, is one of the strongest predictors of depression [7].
poor self-rated health and depression in people Besides depression, diabetes distress (the neg-
with established DPN [2–4]. ative affective or emotional experience resulting
Numerous neurobiological similarities tie dia- from living with the demands of diabetes) and
betic neuropathies, pain, depression, and anxiety anxiety are also common in diabetes [12]. In fact,
together—these threads include the neuroendo- elevated anxiety symptoms were found to be
crine system, autonomic system, and the inflam- more prevalent than depressive symptoms, with
32% of people with diabetes reporting elevated
symptoms of anxiety and 22% for elevated symp-
L. Vileikyte (*)
Diabetes, Endocrinology and Gastroenterology, toms of depression, using the Hospital Anxiety
University of Manchester, Manchester, UK and Depression Scale [13]. The observed diabe-
Department of Dermatology, University of Miami, tes and anxiety relationship is consistent with
Miami, FL, USA accumulating research linking chronic pain and
e-mail: lvileikyte@med.miami.edu anxiety; this pattern mirrors but exceeds the pat-
F. Pouwer tern examining depression [14]. These observa-
Department of Psychology, Steno Diabetes Center tions might be of potential importance to
Odense, University of Southern Denmark, managing painful DPN, as commonly used medi-
Odense, Denmark

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 221
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_13
222 L. Vileikyte and F. Pouwer

cations are potent mood modulators with distinct have traditionally focused on painful DPN. One
class effects on anxiety and depression. of the earliest, yet probably most comprehensive
Accordingly, we discuss studies that examined assessments to-date of neuropathic pain burden,
the differing contributors to anxiety and depres- was conducted by Gore et al. [20]. This commu-
sive symptoms as well as different predictors of nity based, cross-sectional self-report survey
analgesic response to duloxetine and pregabalin described pain severity, as measured by the
in persons with painful DPN [15, 16]. Again, we Modified Brief Pain Inventory-DPN [21], its
emphasize the importance of distinguishing gen- interference with function, as well as prescription
eralized distress (depression/anxiety) from dia- medication use and treatment satisfaction among
betes/DPN-specific distress, a psychological patients with painful DPN.  The burden of pain
concept although related to, but not interchange- was reported to be considerable, resulting in a
able with, depression or anxiety [12, 17]. A grow- persistent discomfort despite polypharmacy and
ing number of researchers studying DPN favor high resource use, and leading to limitations in
disease-specific over psychiatric definitions of daily activities and poor satisfaction with treat-
emotional distress. Using a diabetic neuropathy ments that were often deemed to be inappropri-
and depression model we illustrate how DPN-­ ate. Individuals with painful DPN reported
specific distress mediates the association between significant impairment in physical and mental
DPN severity and depression [2, 3]. Another functioning as compared to the general US popu-
emerging line of inquiry examines DPN-specific lation, although the differences in mental func-
fears such as fear of pain increase and/or fear of tioning were less pronounced than those observed
falling and their associations with greater disabil- in physical functioning. A qualitative study of
ity and reduced quality of life (QoL) in persons persons affected by painful DPN [22] identified
with DPN [18, 19]. Pertaining to this, by compar- four major areas of impact: (1) physical function,
ing and contrasting the performance of generic i.e. walking, exercise, energy, standing, balance,
and DPN-specific QoL instruments, we make a bending, and mobility; (2) daily life, i.e. produc-
case for the need to consider DPN-specific mea- tivity, recreational activities, work, enjoyment,
sures when identifying and managing the psy- focus, and chores; (3) social/psychological, i.e.
chosocial problems surrounding this devastating anxiety, friends/family, irritability, depression,
complication of diabetes, both in clinical care and fear; and (4) sleep, i.e. falling asleep, waking
and in clinical research projects. Finally, we pro- in the night and not feeling rested upon
vide a critical overview of the paucity of psycho- awakening.
social interventions conducted in this area and In a study by Zelman et  al. [23], individuals
emphasize the need for an integrated, bio-­ with painful DPN reported impaired sleep rela-
psychosocial approach to the management of tive to the US population norms and the Medical
DPN. This chapter concludes by discussing psy- Outcomes Study (MOS) chronic patient sample
chological treatments and interventions that on both, the sleep problem index and all six sleep
could potentially assist in the management of attribute scales. Hierarchical regression indicated
psychosocial hardships experienced by people that older age, higher average daily pain, and
with DPN. higher levels of anxiety and depression were each
significantly associated with, and collectively
accounted for, 47% of variance in the MOS sleep
2 The Impact of DPN problem index. More recently, Naranjo et al. [24]
on Physical and Mental in a cross-sectional study compared sleep charac-
Functioning, and QoL teristics in type 2 diabetes patients with and with-
out painful DPN and investigated the relationship
Although DPN is a multifaceted complication of of sensory phenotypes, anxiety, and depression
diabetes, studies examining the impact of DPN with sleep quality. Persons with painful DPN
on individuals’ physical and mental functioning reported significantly more sleep disturbances
Psychosocial Aspects of Diabetic Neuropathy: From Description to Interventions 223

than the control subjects in all dimensions of the of them using anticonvulsants or antidepressants.
MOS sleep scale. Higher scores in anxiety or Similarly, a French study confirmed the notion
depression, greater intensity of pain or a higher that persons with painful DPN have poorer QoL
score in the paroxysmal pain phenotype were and more sleep disturbances, anxiety, and depres-
associated with lower sleep quality in individuals sion than those without pain, with only 39% of
with painful DPN. As shown by Tolle et al. [25], the persons receiving appropriate treatment for
inadequate pain control and severe sleep depriva- their painful DPN [28]. Another study conducted
tion resulted in over 60% of persons with painful in Wales indicated that the severity of painful
DPN who worked either part or full-time report- DPN symptoms predicts poor health-related util-
ing either missing work or decreased work pro- ity and decreased QoL [29]. Happich et al. [30]
ductivity. Worse yet, in this report, pain intensity described the health-related QoL, using generic
disrupted substantially employment status, with and disease-specific questionnaires, the resource
nearly 20% of persons with painful DPN report- utilization, and annual costs associated with DPN
ing job loss or early retirement. A subsequent in Germany. In this retrospective, observational
prospective study [26] confirming and expanding study of persons with type 1 and type 2 diabetes
upon the literature, showed a significantly worse and DPN, the majority of them were severely
trajectory of outcomes over time and long-term impaired with regard to general physical health.
increased total costs for individuals with painful Disease-specific QoL decreased continuously
DPN relative to those without painful DPN and with increasing DPN severity. In accordance,
control subjects. The painful DPN group not only costs associated with DPN increased with the
reported significantly lower levels of physical progression of DPN, with costs from the societal
functioning, but their physical functioning scores perspective increasing about 50-fold from the
decreased at a significantly faster rate over a lowest severity stage (those with sensory-motor
3-year period relative to other groups. In addi- neuropathy without symptoms) to persons with
tion, those with painful DPN reported signifi- lower extremity amputation. Canadian research-
cantly lower work productivity and activity, ers [31] have demonstrated that in persons with
greater resource use, and higher total 3-year per-­ longstanding type 1 diabetes, compared to other
person costs. Resource use and lost productivity complications, DPN has the greatest association
due to health were monetized separately for each with both diabetes distress and depression inde-
year and then totaled for the 3-year period. pendent of clinical and lifestyle confounding
Although the costs were generally comparable variables. A large Danish study estimated the
between the diabetes without painful DPN and prevalence of painless and painful DPN, impor-
control groups, those with painful DPN were tant risk factors, and the association with mental
estimated to have significantly higher direct and health in persons with recently diagnosed type 2
total costs than the other groups in each year of diabetes [32]. The group demonstrated that pos-
the study. The main contributors to increased cost sible painless and painful DPN were indepen-
in the painful DPN group were the increased dently and additively associated with lower QoL,
number of physician visits and hospitalizations. poorer sleep, and elevated symptoms of depres-
The impact of DPN on individuals’ physical sion and anxiety.
and mental function originally reported by US These findings are important given that health-
researchers has recently received much attention care costs are significantly higher for DPN
in other countries. A study of hospital outpatient patients with depression or anxiety relative to
clinics across Belgium [27] showed that both those without such comorbid disorders [33].
painless and painful DPN are common, espe- Several recent systematic reviews [34, 35] of psy-
cially in people with type 2 diabetes. Despite its chosocial factors related to painful DPN have
profound negative impact on QoL, painful DPN also reaffirmed the results of individual studies
remains undertreated with only half of individu- indicating that depression, anxiety, sleep quality,
als receiving analgesic treatment, and only 28% and quality of life are the most studied variables
224 L. Vileikyte and F. Pouwer

in relation to pain outcomes and are consistently effect. Therefore, while there was no significant
associated with pain intensity. difference in analgesic efficacy between amitrip-
tyline, duloxetine, and pregabalin, there were sig-
nificant differences in the secondary parameters,
3 The Effects of Pain which are of relevance when deciding the opti-
Medication on Psychosocial mal treatment for painful DPN.  Somewhat sur-
Outcomes: Sleep and Mental prisingly, this study [36] found no significant
Health improvements in mental health as assessed by the
Short Form (SF)-36 which is unexpected given
Painful DPN is difficult to treat, with treatment that all three treatments (pregabalin for general-
regimens often inadequate for pain control and ized anxiety disorder and duloxetine and amitrip-
limited by side effects and drug intolerance. tyline for depression) are indicated for affective
Secondary parameters, such as quality of sleep disorders. Moreover, as evident from the studies
and mood, may also be important for successful reviewed in this chapter, painful DPN is associ-
painful DPN management. A randomized, ated both with depression and anxiety. Boyle and
placebo-­ controlled trial study by Boyle et  al. colleagues therefore proposed that the (SF)-36
compared the analgesic efficacy of pregabalin, measure is not sensitive enough to assess changes
amitriptyline, and duloxetine, and their effect on in mood and that more specific measures might
sleep, daytime functioning, and quality of life in be more appropriate to detect subtle changes in
patients with painful DPN [36]. All medications mood state over time.
reduced pain when compared with placebo, with
pain ratings showing 50% improvement, in line
with previous studies. However, no treatment was 4 The Role of Psychosocial
superior to any other. Daytime performance mea- Factors in Pain Experience
sures showed no evidence of cognitive impair- and Response to Treatment
ment during treatment. For sleep, pregabalin
improved sleep continuity, whereas duloxetine While the impact of painful DPN on individuals’
increased wake and reduced total sleep time. The psychosocial functioning is well researched, little
beneficial effects of pregabalin on pain-related is known about the role of psychosocial factors in
sleep interference were documented in a review shaping pain experience and persons’ response to
of clinical trials [37] suggesting that, in addition treatment. A study by Otto et al. [39] is one of the
to its analgesic properties, pregabalin may affect earliest reports that examined the impact of indi-
pain perception indirectly, that is, through viduals’ mental and physical functioning, as
improving sleep quality. Indeed, a post-hoc anal- measured by the SF-36, on their response to
ysis of data pooled from placebo-controlled trials treatment for painful DPN. The researchers dem-
of pregabalin in persons with painful DPN using onstrated that individuals’ physical, social and
path modeling showed a high degree of associa- emotional status predicts their response to treat-
tion between improvements in sleep and pain ment after controlling for demographic variables,
relief. Overall, these data suggest that the pres- pain duration, and intensity. The authors there-
ence of comorbid sleep disturbance in persons fore suggested that psychosocial factors might
with painful DPN might, in part, predict substan- provide a useful guide in selecting the popula-
tial pain relief in response to pregabalin treatment tions most responsive to treatment for painful
[38]. The sleep fragmentation with duloxetine, as DPN. Marchettini et al. [16] using data from the
observed in the study by Boyle et  al. [36], is COMBO-DPN study, a multinational clinical
somewhat concerning. It is widely believed that trial in painful DPN, investigated whether there
poor sleep quality may worsen pain, and although are different predictors of analgesic response
duloxetine has good analgesic efficacy, its effec- between duloxetine and pregabalin in painful
tiveness may be limited by this physiological DPN.  There were no significant interactions
Psychosocial Aspects of Diabetic Neuropathy: From Description to Interventions 225

between treatment for subgroups by demo- bility accounted for significant variance in pain-
graphic, baseline pain characteristics, glycemic ful DPN experience. However, the variance
control or presence of comorbidities, and con- accounted for by psychological variables was
comitant medication use. The only significant less than that accounted for by pain intensity.
interaction with treatment was observed in the These results suggest that psychological flexibil-
mood symptom subgroups with a larger pain ity may play a smaller role, relative to pain inten-
reduction in duloxetine-treated persons having sity, in the context of painful DPN as compared
no mood symptoms. Selvarajah et al. [15] exam- to the larger populations of chronic, mostly mus-
ined the contribution of demographic, social, culoskeletal, pain. A study by Lewko et al. [42]
clinical, and psychological factors (Pain showed that poorer QoL in people with DPN is
Acceptance and Pain Catastrophizing) to anxiety related to their lower levels of illness acceptance.
and depressive symptoms in persons with painful Among psychosocial factors negatively affecting
DPN. This study found a high prevalence of emo- illness acceptance were feelings of being a bur-
tional distress with over 50% of individuals expe- den to their family and friends and the belief that
riencing symptoms of anxiety/depression. people in their company are made anxious by the
Multiple regression analysis showed that cata- person’s illness.
strophic thinking is an independent contributor to Vileikyte and Gonzalez [43] examined the
greater symptoms of anxiety and depression. role of psychological variables (generalized dis-
Being young, single, and unemployed signifi- tress, personality traits: extroversion, agreeable-
cantly contributed to greater anxiety symptoms ness, openness, conscientiousness, and
while pain-related restrictions of quality of life neuroticism, and DPN-specific cognitions) and
were associated with greater depression symp- sleep deprivation in generating and sustaining
tom scores. The report therefore highlighted the painful DPN. Both cross-sectional and longitudi-
differing independent contributors to anxiety and nal models were largely consistent, with depres-
depressive symptoms which are largely based on sive and, in particular, anxiety symptoms
an individual’s circumstances and experience. independently associated with more severe pain
Another study investigated the relationship of over time. Sleep impairment and pain unpredict-
pain catastrophizing with disability and QoL in ability cognitions were also predictive of
persons with painful DPN [40]. Additionally, the increases in pain. The personality trait “neuroti-
mediating roles of physical activity and/or cism” was the only personality characteristic
decline in physical activity were explored. The with significant cross-sectional and longitudinal
findings emphasize the role of catastrophic think- relationships to painful DPN, although its effects
ing about pain that was associated with an were confounded with anxious and depressive
increased disability and decreased QoL as well as symptoms. The results also indicated the substan-
with a perceived decline in physical activity, tial chronicity of pain, with baseline pain severity
which in turn, mediated the association between explaining 62% of the variance in painful symp-
catastrophizing and disability and QoL.  Kioskli toms at 18 months. Change-based analyses, con-
et  al. [41] investigated whether psychological trolling for baseline pain severity, showed that
flexibility is potentially beneficial for people with younger age and higher levels of anxiety were the
painful DPN. Psychological flexibility is a model only significant predictors of increments in pain
of well-being and performance that includes sev- over time. These data suggest that although DPN
eral related processes: acceptance, committed pain experience is commonly accompanied by
action, cognitive defusion or ability to separate depression, sleep impairment, and negative cog-
one’s thoughts from events as they are directly nitive appraisals, these factors are unlikely to be
experienced and self-as-context that is not based causal contributors or symptom amplifiers to
upon self-evaluations and is separate from one’s pain. The most important predictor of pain over
thoughts and feelings. In regression analyses, the time is baseline pain severity, although anxiety
four variables representing psychological flexi- may also play a causal role. These findings once
226 L. Vileikyte and F. Pouwer

again emphasize the intimate linkage between independently associated with higher depression
painful DPN and anxiety. The observed painful symptoms and together accounted for the rela-
DPN and anxiety relationship is consistent with tionship between the clinical measures of DPN
accumulating research linking chronic pain and severity and depression scores. The association
anxiety; this pattern mirrors but exceeds the pat- between DPN symptoms and depression was par-
tern for research examining depression and tially mediated by two sets of DPN-specific psy-
chronic pain [14]. More research is still needed, chosocial factors: (1) restrictions in ADL and
particularly that which uses prospective methods, diminished self-worth (self-perception as a fam-
to delineate the temporal primacy and the mecha- ily burden) and (2) illness cognition, or the per-
nisms accounting for the enduring association ceptions of neuropathic pain unpredictability and
between painful DPN and anxiety. the lack of treatment control. The longitudinal
findings of Vileikyte et al. [3] were largely con-
sistent with their cross-sectional observations
5 Pathways Linking DPN and demonstrated that more severe DPN at base-
and Its Symptoms line is associated with worsening depression
to Depression and Anxiety symptoms over 18-months of follow-up. While
neuropathic pain contributed to depression, it
Several earlier reports by Vileikyte et  al. exam- was particularly DPN-postural instability and its
ined both cross-sectionally [2] and prospectively psychosocial consequences that dominated this
[3] the associations between the objective tests of relationship over time. DPN-postural instability
DPN severity and depression symptoms and continued to be a significant predictor of depres-
explored the potential physical and psychosocial sion symptoms with the hypothesized psychoso-
mediators of this association. The selection of cial mediators in the equation, thereby suggesting
psychosocial factors was based on existing that it is related to depression through mecha-
hypotheses linking physical illness to depression. nisms that are not fully accounted for by these
For example, Williamson [44] postulated that studies. It has been proposed, for example, that
illness-­related functional disability and restric- perceived postural instability represents not only
tions in activities of daily living (ADL) can balance dysfunction but also other domains of
directly contribute to the development of depres- vulnerability, such as decreased physiological
sion. Heidrich et  al. [45] argue that the role of reserve and psychosocial factors including poor
ADL restrictions in generating depression may self-efficacy with walking and fear of falling
depend on the extent to which being unable to [47]. These are established risk factors for func-
perform daily activities has a negative impact on tional decline [48] and might have provided addi-
sense of self. Furthermore, it has been proposed tional linkages of DPN-postural instability to
that shared negative cognitive schemata underly- depression symptoms.
ing physical illness and depression may create The emergence of DPN-postural instability as
the possibility of a psychological path linking the symptom that is most strongly associated
physical illness to depressive symptoms: both are with depression merits attention from clinicians,
perceived as chronic, uncontrollable and poten- especially as the preliminary evidence presented
tially having serious consequences [46]. The in a systematic review [49] suggests that people
cross-sectional results of the study by Vileikyte with type 2 diabetes and DPN can improve their
et al. [2] demonstrated that DPN severity is sig- balance and walking following exercise training
nificantly associated with higher levels of depres- interventions, such as lower limb strengthening,
sion symptoms, with DPN symptoms and balance practice, aerobic exercise, walking pro-
DPN-specific psychosocial factors accounting grams, and Tai Chi. We demonstrated that DPN-­
for nearly half of the variance in depression postural instability is common, with nearly a
scores. Neuropathic pain, DPN-postural instabil- quarter of DPN patients reporting balance prob-
ity and reduced feeling in the feet were each lems as present either most or all the time [50].
Psychosocial Aspects of Diabetic Neuropathy: From Description to Interventions 227

Unfortunately, balance deficits may be over- consider a multifaceted approach to the manage-
looked by clinicians, as patients often do not ment of individuals with DPN-postural instabil-
report balance concerns during medical consulta- ity, including a biomechanical component
tions owing to the perception that these are an (physical adaptation to postural instability to
indicator of diminishing self-resources-a sign of improve safety) and a psychosocial component
premature aging rather than illness-related (strategies aimed at increasing participation in
­disability [50]. Importantly, these patients have a daily activities and enhanced perceptions of com-
~20-fold greater risk of falling compared to aged-­ petence in important social roles). The impor-
matched controls [51]. The consequences of fall- tance of postural instability as a symptom of
ing, however, are not only physical but also DPN should receive attention in the education of
psychological, such as poor self-efficacy with both persons with DPN and their healthcare
walking and fear of falling [47]. This highlights providers.
the negative spiral between DPN, postural insta-
bility, falls, and psychological distress. Although
it is acknowledged that psychological factors 6 Generalized Anxiety Vs.
such as fear of falling can powerfully impact Specific Fears
upon individuals’ confidence with walking,
resulting in decreased participation in activities Persons with chronic diseases commonly report
of daily living, elevated depression symptoms fears of illness or symptoms recurring or worsen-
and diminished overall QoL, no psychological ing that have been addressed from several view-
interventions to date have been performed in peo- points: an illness-specific perspective (e.g., fear
ple with DPN-postural instability. Nonetheless, of hypoglycemia), a generic illness perspective
some positive effects on reducing falls by means (e.g., fear of progression), and a psychiatric per-
of psychological interventions have been spective (illness anxiety disorder and somatic
achieved in high-risk older adults. One multifac- symptom disorder) [54]. A growing number of
torial intervention that did significantly reduce researchers, including those studying DPN, favor
the risk of falling in at-risk elderly patients uti- disease-specific over psychiatric definitions of
lized a cognitive behavioral skills learning illness anxiety. Fear of amputation, for example,
approach [52]. The intervention program aimed is emerging as a predominant emotion in people
to improve self-efficacy for preventing falls, with diabetic foot ulceration and is a powerful
encourage behavior change, and provide educa- determinant of preventive foot self-care actions
tion about risk management. Over 14 months of [55–57]. Similarly, people with DPN-postural
follow-up, intervention group participants expe- instability experience psychological conse-
rienced a clinically meaningful 31% reduction in quences such as poor self-efficacy with walking
falls. Furthermore, those in the intervention and fear of falling [58]. A group of Dutch investi-
group were more confident in their ability to gators conducted a qualitative study that identi-
avoid a fall during a variety of functional daily fied a series of diabetes-specific fears experienced
living tasks and used more protective behavioral by people with DPN suffering from pain: fear of
practices than control participants. Similar meth- hypoglycemia, fear of pain increase, fear of total
odologies adapted for patients with DPN could exhaustion, fear of physical injury, fear of falling,
be effective and deserve empirical evaluation. fear of loss of identity, and fear of negative evalu-
Considering this discussion, clinicians should ation by others [18]. Subsequently, the same
therefore address postural instability as a key research group subjected their qualitative obser-
symptom when assessing persons with DPN, vations to quantitative examination [19]. In a
especially as his/her perception of their postural cross-sectional study of persons with painful
instability appears to be an adequate indicator of DPN, all fears were independently associated
the actual balance impairment [53]. Pending a both with reduced QoL and with greater disabil-
definitive intervention study, clinicians should ity, after adjusting for appropriate control vari-
228 L. Vileikyte and F. Pouwer

ables. Linear regression models including all whether DPN is measured by clinical tests of
fears and confounders showed that pain intensity, neurological dysfunction [60] or by patient
pain duration, and fear of falling were signifi- somatic experience with DPN [36, 61]. For
cantly associated with reduced QoL while pain example, Padua et  al. [60] showed that DPN
intensity, male gender and fear of falling were severity, as measured by clinical and neuro-­
significantly associated with greater disability. physiological tests, correlated with physical but
The predominance of fear of falling in persons not mental aspects of SF-36. The authors of this
suffering from painful DPN is an interesting find- paper therefore concluded that DPN is “strictly
ing that requires further investigation. It is plau-
related to the physical aspects of the patients’
sible that people with painful DPN are also quality of life, and not the mental aspects”.
experiencing postural instability related to their Similarly, a prospective investigation reported a
diabetic neuropathy, as it is well-established thattemporal link between the development of pain-
DPN-postural instability is associated with fear ful DPN, foot ulceration and amputations and a
of falling and reduced physical activity [58]. It subsequent decline in SF-36 physical but not
could also be possible that medications used to mental function [61]. More recently, in a large
alleviate pain are negatively impacting the peo- cross-sectional study examining the associations
ples’ balance through their recognized effects on between microvascular and macrovascular com-
the central nervous system [59]. Nonetheless, it isplications of diabetes and health status, DPN was
an interesting report confirming that persons with associated with the greatest reduction in physical
painful DPN suffer from a variety of fears that but not mental component score as measured by
could be tackled by a carefully designed psycho-­ SF-36 [62]. However, the conclusion that DPN
educational intervention, thereby improving and its clinical manifestations do not impact
physical and psychosocial well-being of these mental function is not necessarily valid, espe-
individuals. cially as a number of studies reviewed in this
chapter reported on a significant association
between DPN and elevated levels of both gener-
7 Measuring QoL in DPN: alized (anxiety/depression) and DPN-specific
Generic, DPN-Specific or emotional distress. The alternative explanation
Combined Approach? could be that the generic measures of health sta-
tus are less sensitive and thus do not capture ade-
7.1 Limitations of the Generic quately neither generalized nor DPN-specific
Health status/QoL emotional disturbance. Vileikyte et al. [50] com-
Instruments pared the performance of the SF-12 and a neu-
ropathy and foot ulcer-specific quality of life
The main reason for using a generic measure of questionnaire, the NeuroQoL, providing support
health status or QoL is that the potential impact for the latter. This study demonstrated that while
of different diseases can be compared. For more the mental functioning scale from the SF-12 was
refined assessment of the impact of a certain con- not associated with DPN severity, a DPN-­
dition, validated disease-specific assessment emotional distress scale from the NeuroQoL
tools are usually the preferred option. Cross-­ showed a strong association with DPN severity
sectional and longitudinal studies in DPN that and was the most important link between DPN
employed the generic health status/QoL instru- and diminished QoL.  The results of a compre-
ments, such as the most commonly used, SF-36, hensive review that assessed the impact of neuro-
revealed a common pattern, that is, that DPN pathic pain on QoL supported these observations.
affects health predominantly in the domain of It provided evidence that the impact of neuro-
physical functioning: the impact of DPN on men- pathic pain varies in part as a function of the QoL
tal functioning was non-significant in most of domain being considered, with the condition-­
these reports. This pattern holds regardless specific QoL measures being more sensitive to
Psychosocial Aspects of Diabetic Neuropathy: From Description to Interventions 229

the effects of neuropathic pain than the generic with various peripheral neuropathies including
instruments of physical and especially mental DPN (PN-QOL-97). This instrument consists of
functioning [63]. Taken together, it would seem 97 items and encompasses several scales with
appropriate to supplement the generic tools of varying specificity of assessment, such as an
health status with validated anxiety and depres- item on overall health from the generic
sion scales when assessing the levels of EuroQoL, the entire SF-36 along with items
­generalized distress while considering condition-
measuring symptoms specific to peripheral neu-
specific questionnaires to study DPN-specific ropathies and dysfunction. The 47-item Norfolk
emotional distress. QoL-DPN, in addition to the generic health sta-
Another inherent limitation of the generic tus and general information items includes a
instruments is that their content was imposed bycomprehensive measure of DPN symptoms and
the investigators and did not emerge from inter-the specific ways these symptoms impact on
views with people affected by DPN.  Although ADL [66]. The important limitation of this
the generic measures allow the comparisons instrument is that it does not incorporate DPN-
between different disease states at the most specific emotional distress which is central to
abstract level, they lack specificity, that is, they
DPN experience. The 35-item NeuroQoL [50],
do not describe the specific clinical features of
derived from the discussions with focus groups
DPN and the ways these features impair of persons with DPN and developed in collabo-
QoL. Therefore, the findings from these studies ration neuropathy and psychology experts, is a
leave a gap between DPN as abstractly defined hierarchically organized scale that assesses an
and the person’s experience of DPN that is essen-
individual’s subjective reports of functioning
tial for framing effective interventions. and QoL in six specific domains. Following the
It is also important to note that although hierarchical model [67], the base of each domain
appraisals of mental, physical and social func- is assessed with items that measure DPN-
tioning are important in QoL decision-making, specific somatic experiences (pain, unsteadi-
they are not direct measures of QoL. Individualsness, and symptoms of reduced feeling in the
can report similar levels of dysfunction and differ
feet), DPN-specific social and personal dys-
in their subjective judgments as to the impact of
function, and emotional states. The two final
these functional impairments on their items in the scale complete the hierarchical
QoL. Therefore, QoL is what it appears to be: “aapproach by requesting that the patients appraise
subjective judgment of quality of one’s life andthe impact of DPN on their QoL and provide an
overall QoL evaluation. The psychometric anal-
not a measure of physical or cognitive function, a
report of emotional state, or a measure of a yses that compared the performance of the
patient’s integration into a social network” [64].
NeuroQoL to the SF-12 demonstrated that the
Such a definition of QoL necessitates therefore NeuroQoL was more strongly associated with
the inclusion of items directly asking respondents
DPN severity (criterion validity), more fully
to subjectively evaluate their QoL. mediated the relationship of DPN with QoL
(construct validity) and significantly increased
explained variance in QoL judgment over the
7.2 The Shift from Generic SF-12 (incremental validity) [50].
to DPN-Specific QoL Following the development of DPN-specific
Assessments QoL instruments, a number of systematic
reviews and DPN expert guidelines subjected
In an attempt to overcome the limitations of the these measures to scientific scrutiny. Two sys-
generic instruments, three DPN-specific QoL tematic reviews of patient reported outcomes
measures have been developed. The health-­ (PROs) concluded that although the DPN-
related QoL measure by Vickrey et al. [65] was specific tools were better than generic in quanti-
developed for the assessment of QoL in persons fying temporal changes in QoL and showed
230 L. Vileikyte and F. Pouwer

greater sensitivity to neuropathy severity, no one position statement regarding psychosocial care
measure was identified as a “gold standard” for for people with diabetes by the American
assessing QoL in DPN.  The authors therefore Diabetes Association (ADA) therefore recom-
recommended that the instrument selection mended that in general: “…psychosocial care
should depend on the clinical and social context should be integrated with collaborative, patient-­
of the assessment [68, 69]. An international centered medical care and provided to all people
workshop of neurology experts on the selection with diabetes, with the goals of optimizing health
of outcome measures for clinical trials in periph- outcomes and health-related quality of life” [72,
eral neuropathy recommended the use of 73]. A key element of collaborative, patient-­
PN-QOL-97 and the NeuroQoL [70], while a centered diabetes care is that levels of diabetes
recent Position Statement by the American distress, depression, anxiety, and disordered eat-
Diabetes Association covering diabetic neuropa- ing are monitored and discussed. This should be
thies proposed the Norfolk QoL-DPN and the done using patient-appropriate validated tools “at
NeuroQoL for QoL assessment in DPN [1]. the initial visit, at periodic intervals, and when
Examination by neurology experts of the content there is a change in disease, treatment, or life cir-
validity of symptom-based measures for dia- cumstance” [72]. In collaborative or stepped
betic, chemotherapy, and HIV peripheral neu- care, treatment for depression is intensified if the
ropathies concluded that given significant symptoms still persist [74].
overlap in symptoms between neuropathy etiol- In the ADA position statement [72], a specific
ogy, a measure with content validity for multiple paragraph focuses on diabetes complications and
neuropathies with supplemental disease-specific functional limitations. People with diabetes com-
modules could be of great value in the develop- plications, including neuropathy, foot ulcers or
ment of disease-modifying treatments for limb amputation not only experience depression
peripheral neuropathies [71]. Therefore, it would and anxiety, but also have lower overall physical
appear that in order to advance this complex function and impaired QoL.  This can result in
area, it would seem appropriate that the develop- reduced autonomy and serious role impairments
ment and/or refinement of QoL instruments and [72, 75]. It is therefore recommended that in dia-
their critical appraisals were conducted by a betes care settings the levels of chronic pain asso-
team consisting of persons with DPN, experts ciated with diabetes complications and the impact
both in DPN and in developing PRO measures. of painful DPN or discomfort on QoL are moni-
Additional research on the psychometric charac- tored. After a discussion of questionnaire scores,
teristics of existing measures is warranted, these appropriate pain management therapies should
future studies should follow rigorous guidelines be offered, including a consultation with a mental
for reporting on PROs [69]. health provider for pain self-management strate-
gies [72].
In their comment on the above ADA position
8 How to Manage statement, Snoek et al. propose that it is key to
Psychological Problems emphasize that the absence of a serious mental
of Persons with DPN health problem is not the same as emotional
in Clinical Care? well-­being or “a good quality of life” [76].
Another important remark by these authors is
As described in this chapter, psychological prob- that the use of screening tools should never
lems are common in people with diabetes, espe- become a routine procedure where health care
cially in those with diabetes complications, such providers are simply “ticking the box.” The
as neuropathy. Diabetes distress, depression, and score(s) on a questionnaire should always be
disordered eating can not only seriously impair regarded as a tool that can help to facilitate and
quality of life, but also negatively impact self-­ improve the clinical conversation. This is impor-
care behaviors and long-term outcomes [8]. A tant, as a questionnaire score that is indicative of
Psychosocial Aspects of Diabetic Neuropathy: From Description to Interventions 231

a mental health problem does not by definition (CBT, 59 studies). The systematic review fur-
equal a need for psychiatric or psychological thermore has included eight studies that tested
care. Diabetes health care teams should be behavioral therapy (BT), and five studies that
trained to carefully explore this with an individ- investigated Acceptance and Commitment
ual with diabetes. If needed, culturally accept- Therapy (ACT). The results of the systematic
able, affordable evidence-based treatment review suggest that there is moderate-­quality
options should be offered in response to the evidence showing that individuals treated with
identified psychosocial needs [76]. CBT experience less pain and distress by the
Many questionnaires can be used to support end of the treatment and 6–12  months later,
clinical diabetes care. In the ADA position state- compared to those who had received no treat-
ment, several questionnaires are recommended, ment for their pain, though effect sizes were
including the Problem Areas in Diabetes Survey relatively small. Those in the intervention
[77] or Diabetes Distress Scale [78] for diabetes groups also reported less disability on average
distress, Patient Health Questionnaire (PHQ)-9 (low-quality evidence). Furthermore, moder-
or Beck Depression Inventory (BDI-II) for ate-quality evidence showed that CBT reduced
depression [79]. The conversation after the pain, disability, and distress in comparison with
assessment can lead to further evaluation or a individuals receiving a non-psychological
structured psychiatric diagnostic interview. For intervention for their pain, such as an exercise
the assessment of chronic pain, it is recom- program, or education about how to manage
mended to use the Short-form McGill Pain pain, but here too, the effect sizes were rela-
Questionnaire (SF-MPQ-2) [80]. This tool can tively small. At 6–12  months follow-up, pain
also be used to determine in clinical care whether and distress were still significantly lower (mod-
the treatment for painful DPN is effective. It is erate-quality evidence), while disability levels
also important to emphasize that pain is by defi- did not differ between CBT and non-psycho-
nition a subjective, personal experience and that logical treatment (low-quality evidence) [82].
emotional difficulties such as loneliness, stress or Another systematic review with meta-analy-
depression can definitely negatively impact the sis specifically focused on the effectiveness of
way people experience and act as symptom different psychological interventions on pain
amplifiers of pain [81]. and related outcomes in adults with diabetic
peripheral neuropathy [83]. In this review, nine
trials were included, showing that in the short-
9 Psychological Treatment term, psychological therapies had a large posi-
Options for People tive effect on pain severity, and a small effect
with Painful DPN on pain interference. In adults with DPN, psy-
chological interventions were concluded to
Different psychological interventions have have a moderate effect on depressive symp-
been designed to aid people to improve the way toms. When the effects on medium-term fol-
they cope with chronic pain by changing: (1) low-up assessments were summarized, a large
the way they perceive or appraise pain and (2) effect on pain severity and on pain interference
the way they behave, in order to diminish both was observed, while the effect on depressive
distress and disability. A systematic review of symptoms was moderate. Furthermore, for
75 studies focused on the effectiveness of psy- long-term follow-up assessments, improve-
chological treatments in a number of chronic ments in pain interference, mood, and self-care
pain conditions, such as chronic low back pain, behaviors were reported [83].
rheumatoid arthritis, fibromyalgia, and a mix- Three recent randomized controlled trials
ture of persistent pain conditions [82]. The (RCTs) were not included in the above system-
most commonly investigated psychological atic review. The first small RCT from the USA
intervention was cognitive behavioral therapy (n  =  47) by Higgins et  al. compared CBT with
232 L. Vileikyte and F. Pouwer

diabetes education in people with diabetic neuro- 10 Concluding Remarks


pathic pain [84]. The findings of the study and Directions for Future
rejected the primary hypothesis that those receiv- Research
ing CBT for painful DPN would experience a
larger reduction in pain intensity than those Distal symmetric polyneuropathy, especially
receiving education. The study reported some when accompanied by pain, has a profound and
benefits of CBT relative to education on second- wide-reaching impact on the lives of people, with
ary outcomes, such as, for example, reduction in decrements in physical and mental functioning,
neuropathic pain intensity in the CBT arm. sleep quality, participation in activities of daily
Therefore, CBT is currently the leading living, employment, and overall quality of life.
­psychological treatment for chronic pain, with Unlike painful DPN, postural instability is largely
small to medium effect sizes for CBT relative to underappreciated clinical manifestation of dia-
controls. betic neuropathy. Nonetheless, DPN-postural
One potential new candidate that could help instability is the strongest determinant of depres-
to improve clinical care for people with painful sive symptoms in this population leading to
DPN is Acceptance and Commitment Therapy impaired QoL not only through biomechanical
(ACT). ACT is to be considered as a third wave difficulties but also through associated psycho-
of cognitive behavioral therapies. When ACT is logical distress. There is therefore an unmet need
applied to people with chronic pain, the goal is for the development of multifaceted interventions
to increase valued action in the presence of pain that address both psychological distress and bio-
and also accomplish improvements to function- mechanical challenges experienced by patients
ing. The goal is to increase “psychological flex- with this incapacitating complication of
ibility” through six key processes: (1) diabetes.
acceptance, (2) values-based action, (3) contact The reviewed studies have important implica-
with the present moment, (4) cognitive defu- tions for the care of people with DPN.  They
sion, (5) develop an observer self that can define several possible psychosocial routes link-
change dependent on the context, and (6) com- ing DPN to depression/anxiety and impaired
mitted action in line with important values [85]. QoL thereby providing clinicians with the spe-
ACT was tested in an RCT, where 50 individu- cific points for interventions to alleviate emo-
als with painful DPN were randomized to (1) tional distress and improve QoL. Notably, these
medication alone to manage neuropathic pain reports identify several aspects of DPN-specific
(2) medication and eight sessions of ACT [85]. emotional distress, such as diminished self-worth
The authors reported that ACT increased the and the perceptions of self as a family burden that
level of pain acceptance and reduced pain per- are experienced by persons with painful DPN and
ception. The third study was a single arm pilot with DPN-postural instability. Furthermore, a set
study (n = 30) from the UK [86]. Kioskli et al. of specific fears related to DPN experience is also
[87] were the first to investigate an online ACT prominent and includes fear of falling and fear of
for painful DPN (https://www.act4painonline. worsening of pain, just to mention a few. It is
co.uk), in order to explore whether a larger RCT therefore important to consider not only general-
is warranted. The initial results of this study are ized distress (depression/anxiety) or diabetes dis-
promising. Participants who completed the ther- tress that is associated with the management of
apy reported clinically meaningful effects at glycemia. Importantly, persons’ emotional
post-treatment for 100% of participants for pain responses that are specific to diabetic neuropathy
intensity and pain distress, 67% for depressive should also be assessed and monitored. Although
symptoms, 58% for functional impairment, 42% several DPN-specific instruments capture some
for cognitive fusion, 67% for committed action, of the aspects of neuropathy-related distress [50,
58% for self-as-context, and 42% for pain 56], more work is needed to further develop and
acceptance. refine these scales. The area that warrants strong
Psychosocial Aspects of Diabetic Neuropathy: From Description to Interventions 233

support from researchers, funding organizations, Med. 2020;13:e14383. https://doi.org/10.1111/


dme.14383.
and companies is further standardization of PROs 5. Jain R, Jain S, Raison CL, Maletic V. Painful diabetic
that can be used in clinical care and research of neuropathy is more than pain alone: examining the
people with DPN. The psychometric characteris- role of anxiety and depression as mediators and com-
tics of existing tools should be compared and fur- plicators. Curr Diab Rep. 2011;11:275–84.
6. Lees JG, Fivelman B, Duffy SS, Makker PG,
ther developed in line with rigorous guidelines Perera CJ, Moalem-Taylor G.  Cytokines in neuro-
[85, 86, 88, 89]. New, more effective drugs with pathic pain and associated depression. Mod Trends
fewer side effects should be developed and tested, Pharmacopsychiatry. 2015;30:51–66.
and in these trials, the most optimal PROs should 7. Jacob L, Kostev K. Prevalence of depression in type 2
diabetes patients in German primary care practices. J
be used. Future research should also rigorously Diabetes Complicat. 2016;30:432–7.
test the effectiveness of newer interventions such 8. Pouwer F, Schram MT, Iversen MM, Nouwen A,
as ACT and online ACT or Mindfulness-Based Holt RIG.  How 25 years of psychosocial research
Cognitive Therapy [87, 90]. Offering online has contributed to a better understanding of the
links between depression and diabetes. Diabet Med.
treatment has several advantages, such as easy 2020;37(3):383–92. Epub 2020 Jan 24. PMID:
access, avoidance of waiting lists, people with 31909844. https://doi.org/10.1111/dme.14227.
painful DPN can participate in their own pace, 9. Nouwen A, Adriaanse MC, van Dam K, et  al.
from their own home and during a time of the day European depression in diabetes (EDID) research
consortium. Longitudinal associations between
that they prefer. However, it is also known that depression and diabetes complications: a sys-
drop-out rates also tend to be relatively high. tematic review and meta-analysis. Diabet Med.
Blended care, where online support is offered in 2019;36:1562–72.
combination with a clinical intervention should 10. Black SA, Markides KS, Ray LA.  Depression pre-
dicts increased incidence of adverse health outcomes
also be studied. Future research should also in older Mexican Americans with type 2 diabetes.
investigate how we can become better at prevent- Diabetes Care. 2003;26:2822–8.
ing the development of painful DPN, and how we 11. Lin EH, Rutter CM, Katon W, et al. Depression and
can become better at choosing the right interven- advanced complications of diabetes: a prospective
cohort study. Diabetes Care. 2010;33:264–9.
tion for each individual. Precision medicine stud- 12. Skinner TC, Joensen L, Parkin T.  Twenty-five
ies are needed to learn more about how clinicians years of diabetes distress research. Diabet Med.
can quickly find the most successful or most 2020;37:393–400.
promising intervention for each individual. This 13. Collins MM, Corcoran P, Perry IJ.  Anxiety and
depression symptoms in patients with diabetes.
applies to both pharmacological and psychologi- Diabet Med. 2009;26:153–61.
cal interventions. 14. Asmundson GJ, Katz J.  Understanding the co-­
occurrence of anxiety disorders and chronic pain:
state-of-the-art. Depress Anxiety. 2009;26:888–901.
15. Selvarajah D, Cash T, Sankar A, et al. The contribu-
References tors of emotional distress in painful diabetic neuropa-
thy. Diab Vasc Dis Res. 2014;11:218–25.
1. Pop-Busui R, Boulton AJ, Feldman EL, et  al. 16. Marchettini P, Wilhelm S, Petto H, et al. Are there dif-
Diabetic neuropathy: a position statement by the ferent predictors of analgesic response between anti-
American Diabetes Association. Diabetes Care. depressants and anticonvulsants in painful diabetic
2017;40:136–54. neuropathy? Eur J Pain. 2016;20:472–82.
2. Vileikyte L, Leventhal H, Gonzalez JS, Peyrot M, 17. Snoek FJ, Bremmer MA, Hermanns N.  Constructs
Rubin RR, Ulbrecht JS, Garrow A, Waterman C, of depression and distress in diabetes: time
Cavanagh PR, Boulton AJ.  Diabetic peripheral neu- for an appraisal. Lancet Diabetes Endocrinol.
ropathy and depressive symptoms: the association 2015;3:450–60.
revisited. Diabetes Care. 2005;28:2378–83. 18. Kanera IM, van Laake-Geelen CCM, Ruijgrok JM,
3. Vileikyte L, Peyrot M, Gonzalez JS, et al. Predictors of et al. Living with painful diabetic neuropathy: insights
depressive symptoms in persons with diabetic periph- from focus groups into fears and coping strategies.
eral neuropathy: a longitudinal study. Diabetologia. Psychol Health. 2019;34:84–105.
2009;52:1265–73. 19. Geelen CC, Smeets RJEM, Schmitz S, van den Bergh
4. Hoogendoorn CJ, Gonzalez JS, Schechter CB, et al. JP, Goossens MEJB, Verbunt JA. Anxiety affects dis-
Correlates of self-rated health in people with diabetic ability and quality of life in patients with painful dia-
peripheral neuropathy: a longitudinal study. Diabet betic neuropathy. Eur J Pain. 2017;21:1632–41.
234 L. Vileikyte and F. Pouwer

20. Gore M, Brandenburg NA, Dukes E, Hoffman DL, Tai 33. Boulanger L, Zhao Y, Bao Y, et  al. A retrospec-
KS, Stacey B. Pain severity in diabetic peripheral neu- tive study on the impact of comorbid depression or
ropathy is associated with patient functioning, symp- anxiety on healthcare resource use and costs among
tom levels of anxiety and depression, and sleep. J Pain diabetic neuropathy patients. BMC Health Serv Res.
Symptom Manag. 2005;30:374–85. 2009;9:111.
21. Zelman DC, Gore M, Dukes E, Tai KS, Brandenburg 34. Kioskli K, Scott W, Winkley K, Kylakos S, McCracken
N.  Validation of a modified version of the brief LM. Psychosocial factors in painful diabetic neuropa-
pain inventory for painful diabetic peripheral neu- thy: a systematic review of treatment trials and survey
ropathy. J Pain Symptom Manag. 2005;29(4):401– studies. Pain Med. 2019;20:1756–73.
10. PMID: 15857744. https://doi.org/10.1016/j. 35. Naranjo C, Del Reguero L, Moratalla G, Hercberg
jpainsymman.2004.06.018. M, Valenzuela M, Failde I.  Anxiety, depression and
22. Brod M, Pohlman B, Blum SI, Ramasamy A, Carson sleep disorders in patients with diabetic neuropathic
R. Burden of illness of diabetic peripheral neuropathic pain: a systematic review. Expert Rev Neurother.
pain: a qualitative study. Patient. 2015;8:339–48. 2019;19:1201–9.
23. Zelman DC, Brandenburg NA, Gore M. Sleep impair- 36. Boyle J, Eriksson ME, Gribble L, et al. Randomized,
ment in patients with painful diabetic peripheral neu- placebo-controlled comparison of amitriptyline,
ropathy. Clin J Pain. 2006;22:681–5. duloxetine, and pregabalin in patients with chronic
24. Naranjo C, Dueñas M, Barrera C, Moratalla G, Failde diabetic peripheral neuropathic pain: impact on pain,
I. Sleep characteristics in diabetic patients depending polysomnographic sleep, daytime functioning, and
on the occurrence of neuropathic pain and related fac- quality of life. Diabetes Care. 2012;35:2451–8.
tors. Int J Environ Res Public Health. 2020;17:8125. 37. Roth T, van Seventer R, Murphy TK.  The effect of
25. Tölle T, Xu X, Sadosky AB. Painful diabetic neuropa- pregabalin on pain-related sleep interference in dia-
thy: a cross-sectional survey of health state impair- betic peripheral neuropathy or postherpetic neuralgia:
ment and treatment patterns. J Diabetes Complicat. a review of nine clinical trials. Curr Med Res Opin.
2006;20:26–33. 2010;26:2411–9.
26. daCosta DBM, Cappelleri JC, Joshi AV.  A longitu- 38. Vinik A, Emir B, Parsons B, Cheung R.  Prediction
dinal assessment of painful diabetic peripheral neu- of pregabalin-mediated pain response by severity of
ropathy on health status, productivity, and health care sleep disturbance in patients with painful diabetic
utilization and cost. Pain Med. 2011;12:118–26. neuropathy and post-herpetic neuralgia. Pain Med.
27. Van Acker K, Bouhassira D, De Bacquer D, 2014;15:661–70.
Weiss S, Matthys K, Raemen H, Mathieu C, Colin 39. Otto M, Bach FW, Jensen TS, Sindrup SH.  Health-­
IM. Prevalence and impact on quality of life of periph- related quality of life and its predictive role for anal-
eral neuropathy with or without neuropathic pain in gesic effect in patients with painful polyneuropathy.
type 1 and type 2 diabetic patients attending hospital Eur J Pain. 2007;11:572–8.
outpatients clinics. Diabetes Metab. 2009;35:206–13. 40. Geelen CC, Kindermans HP, van den Bergh JP,
28. Bouhassira D, Letanoux M, Hartemann A.  Chronic Verbunt JA. Perceived physical activity decline as a
pain with neuropathic characteristics in diabetic mediator in the relationship between pain catastro-
patients: a French cross-sectional study. PLoS One. phizing, disability, and quality of life in patients
2013;8:e74195. with painful diabetic neuropathy. Pain Pract.
29. Currie CJ, Poole CD, Woehl A, et al. The health-related 2017;17:320–8.
utility and health-related quality of life of hospital-­ 41. Kioskli K, Winkley K, McCracken LM.  Might psy-
treated subjects with type 1 or type 2 diabetes with chological flexibility processes and acceptance and
particular reference to differing severity of peripheral commitment therapy (ACT) apply in adults with pain-
neuropathy. Diabetologia. 2006;49:2272–80. ful diabetic neuropathy? A cross-sectional survey. J
30. Happich M, John J, Stamenitis S, Clouth J, Polnau Contextual Behav Sci. 2019;13:66–73.
D.  The quality of life and economic burden of 42. Lewko J, Polityńska B, Kochanowicz J, et al. Quality
neuropathy in diabetic patients in Germany in of life and its relationship to the degree of illness
2002—results from the diabetic microvascular com- acceptance in patients with diabetes and peripheral
plications (DIMICO) study. Diabetes Res Clin Pract. diabetic neuropathy. Adv Med Sci. 2007;52(Suppl
2008;81:223–30. 1):144–6.
31. Bai JW, Lovblom LE, Cardinez M, et al. Neuropathy 43. Vileikyte L, Gonzalez JS.  Recognition and manage-
and presence of emotional distress and depression ment of psychosocial issues in diabetic neuropathy.
in longstanding diabetes: results from the Canadian Handb Clin Neurol. 2014;126:195–209.
study of longevity in type 1 diabetes. J Diabetes 44. Williamson GM.  The central role of restricted nor-
Complicat. 2017;31:1318–24. mal activities in adjustment to illness and disabil-
32. Gylfadottir SS, Christensen DH, Nicolaisen SK, et al. ity: a model of depressed affect. Rehabil Psychol.
Diabetic polyneuropathy and pain, prevalence, and 1998;43:327–47.
patient characteristics: a cross-sectional questionnaire 45. Heidrich SM, Forsthoff CA, Ward SE. Psychological
study of 5,514 patients with recently diagnosed type 2 adjustment in adults with cancer: the self as mediator.
diabetes. Pain. 2020;161:574–83. Health Psychol. 1994;13:346–53.
Psychosocial Aspects of Diabetic Neuropathy: From Description to Interventions 235

46. Brownlee S, Leventhal EA, Leventhal H. Regulation, 60. Padua L, Saponara C, Ghirlanda G, et  al. Health-­
self-regulation and regulation of the self in maintain- related quality of life in type 1 diabetic patients and
ing physical health. In: Boekartz M, Pintrich PR, influence of peripheral nerve involvement. Neurol
ZeiDPNer M, editors. Handbook of self-regulation. Sci. 2001;22:239–45.
San Diego, CA: Academic; 2000. p. 369–416. 61. Ahroni JH, Boyko EJ.  Responsiveness of the SF-36
47. McKee KJ, Orbell S, Austin CA, et  al. Fear of fall- among veterans with diabetes mellitus. J Diabetes
ing, falls efficacy, and health outcomes in older Complicat. 2000;14:31–9.
people following hip fracture. Disabil Rehabil. 62. Venkataraman K, Wee HL, Leow MK, et  al.
2002;15(24):327–33. Associations between complications and health-­
48. Lindenberger EC, Landefeld CS, Sands LP, et  al. related quality of life in individuals with diabetes.
Unsteadiness reported by older hospitalized patients Clin Endocrinol. 2013;78:865–73.
predicts functional decline. J Am Geriatr Soc. 63. Jensen MP, Chodroff MJ, Dworkin RH.  The
2003;51:621–6. impact of neuropathic pain on health-related qual-
49. Gu Y, Dennis SM.  Are falls prevention programs ity of life: review and implications. Neurology.
effective at reducing the risk factors for falls in people 2007;68:1178–82.
with type-2 diabetes mellitus and peripheral neuropa- 64. Leventhal H, Colman S.  Quality of life: a process
thy: a systematic review with narrative synthesis. J view. Psychol Health. 1997;12:753–67.
Diabetes Complicat. 2017;31:504–16. 65. Vickrey BG, Hays RD, Beckstrand M. Development
50. Vileikyte L, Peyrot M, Bundy C, et al. The develop- of a health-related quality of life measure for
ment and validation of a neuropathy–and foot ulcer-­ peripheral neuropathy. Neurorehabil Neural Repair.
specific quality of life instrument. Diabetes Care. 2000;14:93–104.
2003;26:2549–55. 66. Vinik EJ, Hayes RP, Oglesby A, et  al. The devel-
51. Schwartz AV, Vittinghoff E, Sellmeyer DE, et  al. opment and validation of the Norfolk QOL-DPN,
Health, aging, and body composition study. Diabetes-­ a new measure of patients’ perception of diabetes
related complications, glycemic control, and falls in and diabetic neuropathy. Diabetes Technol Ther.
older adults. Diabetes Care. 2008;31:391–6. 2005;7:497–508.
52. Clemson L, Cumming RG, Kendig H, Swann M, 67. Fries JF, Sigh G.  The hierarchy of patient out-
Heard R, Taylor K. The effectiveness of a community-­ comes. In: Spilker B, editor. Quality of life and
based program for reducing the incidence of falls in Pharmacoeconomics in clinical trials. 2nd ed.
the elderly: a randomized trial. J Am Geriatr Soc. Philadelphia: Lippincott-Raven; 1996. p. 11–24.
2004;52:1487–94. 68. Hogg FR, Peach G, Price P, Thompson MM,
53. Reeves ND, Brown SJ, Petrovic M, Boulton AJM, Hinchliffe RJ.  Measures of health-related quality
Vileikyte L.  How does self-perceived unsteadiness of life in diabetes-related foot disease: a systematic
influence balance and gait in people with diabetes? review. Diabetologia. 2012;55:552–65.
Preliminary observations. Diabetes Care. 2017;40:e51–2. 69. Bredfeldt C, Altschuler A, Adams AS, Portz JD,
54. Lebel S, Mutsaers B, Tomei C, et al. Health anxiety Bayliss EA.  Patient reported outcomes for dia-
and illness-related fears across diverse chronic ill- betic peripheral neuropathy. J Diabetes Complicat.
nesses: a systematic review on conceptualization, 2015;29:1112–8.
measurement, prevalence, course, and correlates. 70. Merkies IS, Lauria G. 131st ENMC international
PLoS One. 2020;15:e0234124. workshop: selection of outcome measures for periph-
55. Vileikyte L, Pouwer F, Gonzalez JS.  Psychosocial eral neuropathy clinical trials 10-12 December 2004,
research in the diabetic foot: are we making progress? Naarden, the Netherlands. Neuromuscul Disord.
Diabetes Metab Res Rev. 2020;36(Suppl 1):e3257. 2006;16:149–56.
56. Vileikyte L, Gonzalez JS, Leventhal H, et al. Patient 71. Gewandter JS, Burke L, Cavaletti G, et  al. Content
interpretation of neuropathy (PIN) questionnaire: an validity of symptom-based measures for diabetic, che-
instrument for assessment of cognitive and emotional motherapy, and HIV peripheral neuropathy. Muscle
factors associated with foot self-care. Diabetes Care. Nerve. 2017;55:366–72.
2006;29:2617–24. 72. Young-Hyman D, de Groot M, Hill-Briggs F,
57. Wukich DK, Raspovic KM, Suder NC.  Patients with Gonzalez JS, Hood K, Peyrot M.  Psychosocial care
diabetic foot disease fear major lower-­extremity amputa- for people with diabetes: a position statement of
tion more than death. Foot Ankle Spec. 2018;11:17–21. the American Diabetes Association. Diabetes Care.
58. Vileikyte L, Crews RT, Reeves ND.  Psychological 2016;39:2126–40.
and biomechanical aspects of patient adaptation to 73. American Diabetes Association. 5. Facilitating behav-
diabetic neuropathy and foot ulceration. Curr Diab ior change and well-being to improve health out-
Rep. 2017;23(17):109. comes: standards of medical care in diabetes—2021.
59. Tesfaye S, Vileikyte L, Rayman G, et al. Painful dia- Diabetes Care. 2021;44:S53–72.
betic peripheral neuropathy: consensus recommen- 74. Katon WJ, Lin EH, Von Korff M, et al. Collaborative
dations on diagnosis, assessment and management. care for patients with depression and chronic ill-
Diabetes Metab Res Rev. 2011;21:629–38. nesses. N Engl J Med. 2010;363:2611–20.
236 L. Vileikyte and F. Pouwer

75. Argoff CE, Cole BE, Fishbain DA, Irving GA. Diabetic 84. Higgins DM, Heapy AA, Buta E, et  al. A random-
peripheral neuropathic pain: clinical and quality-of-­ ized controlled trial of cognitive behavioral therapy
life issues. Mayo Clin Proc. 2006;81(Suppl):S3–S11. compared with diabetes education for diabetic
76. Snoek FJ, Hermanns N, de Wit M, et al. Psychosocial peripheral neuropathic pain. J Health Psychol.
aspects of Diabetes Study Group of the European 2020;19:1359105320962262.
Association for the Study of Diabetes. Comment on 85. Hughes LS, Clark J, Colclough JA, Dale E, McMillan
Young-Hyman et al. Diabetes Care. 2018;41:e31–2. D.  Acceptance and commitment therapy (ACT) for
77. Kenny E, O'Malley R, Roche K, et al. Diabetes dis- chronic pain: a systematic review and meta-analyses.
tress instruments in adults with type 1 diabetes: a sys- Clin J Pain. 2017;33:552–68.
tematic review using the COSMIN ­(COnsensus-­based 86. Taheri AA, Foroughi AA, Mohammadian Y, et al. The
standards for the selection of health status mea- effectiveness of acceptance and commitment therapy
surement INstruments) checklist. Diabet Med. on pain acceptance and pain perception in patients
2020;24:e14468. with painful diabetic neuropathy: a randomized con-
78. Polonsky WH, Fisher L, Earles J, et  al. Assessing trolled trial. Diabetes Ther. 2020;11:1695–708.
psychosocial distress in diabetes: development 87. Kioskli K, Scott W, Winkley K, Godfrey E,
of the diabetes distress scale. Diabetes Care. McCracken LM. Online acceptance and commitment
2005;28(3):626–31. https://doi.org/10.2337/ therapy for people with painful diabetic neuropathy
diacare.28.3.626. in the United Kingdom: a single-arm feasibility trial.
79. van Dijk SEM, Adriaanse MC, van der Zwaan L, et al. Pain Med. 2020;21:2777–88.
Measurement properties of depression questionnaires 88. U.S Department of Health and Human Services Food
in patients with diabetes: a systematic review. Qual and Drug Administration (FDA). Guidance for indus-
Life Res. 2018;27:1415–30. try: patient reported outcome measures–use in medic-
80. Dworkin RH, Turk DC, Revicki DA, et  al. inal product development to support labelling claims.
Development and initial validation of an expanded 2009. https://www.fda.gov/media/77832/download.
and revised version of the short-form McGill pain Accessed 1 Dec 2018.
questionnaire (SF-MPQ-2). Pain. 2009;144:35–42. 89. Frost MH, Reeve BB, Liepa AM, Stauffer JW, Hays
81. Speight J, Holmes-Truscott E, Hendrieckx C, RD. Mayo/FDA patient-reported outcomes Consensus
Skovlund S, Cooke D.  Assessing the impact of dia- meeting group; what is sufficient evidence for the reli-
betes on quality of life: what have the past 25 years ability and validity of patient-reported outcome mea-
taught us? Diabet Med. 2020;37:483–92. sures? Value Health. 2007;10(Suppl 2):S94–S105.
82. Williams ACC, Fisher E, Hearn L, Eccleston 90. van Laake-Geelen CCM, Smeets RJEM, Quadflieg
C.  Psychological therapies for the management of SPAB, Kleijnen J, Verbunt JA. The effect of exercise
chronic pain (excluding headache) in adults. Cochrane therapy combined with psychological therapy on
Database Syst Rev. 2020;12(8):CD007407. physical activity and quality of life in patients with
83. Racaru S, Sturt J, Celik A. The effects of psychologi- painful diabetic neuropathy: a systematic review.
cal interventions on diabetic peripheral neuropathy: Scand J Pain. 2019;19:433–9.
a systematic review and meta-analysis. Pain Manag
Nurs. 2020;S1524-9042(20):30210–1.
Part II
Pathophysiology
The Genomics of Diabetic
Neuropathy

Abirami Veluchamy, Blair H. Smith,
and David L. Bennett

1 Introduction of neuropathy is ideally captured by a combina-


tion of relevant patient reported outcomes, exam-
For the vast majority of human diseases individ- ination findings and investigations such as nerve
ual susceptibility is to some degree influenced by conduction studies with harmonised definitions.
genetic variation. Understanding the genetics of Given the subjective nature of pain, neuropathic
common disorders such as diabetes mellitus, pain is arguably an even harder phenotype to cap-
heart disease and auto-immune disorders has pro- ture, and important information such as the dura-
vided novel insights into disease pathogenesis, tion, quality and localisation of pain and whether
identified new drug targets and, through combin- this is likely attributable to diabetic neuropathy
ing multiple genetic risk variants into polygenic (e.g., bilateral pain in the feet) is required [3, 4].
risk scores, enabling risk stratification [1]. In this These issues are now beginning to be appreciated
chapter we will discuss how genomics is inform- in large population cohorts such as UK-Biobank,
ing our knowledge of diabetic neuropathy and and such cohorts, allied to advances in sequenc-
also key consequences of diabetic neuropathy ing technology/analytics and the development of
such as neuropathic pain. Compared to our human cellular models, should significantly
understanding of the genomics of diabetes per se advance our understanding of the genomics of
this field (and indeed the genomics of other dia- diabetic neuropathy over the next decade. We
betes complications such as retinopathy and will briefly highlight the broad approaches used
nephropathy) is still very much in its infancy [2]. in human genomics and how they have been
The most important reason for this is the fact that applied to diabetes before focusing on the current
large cohorts with detailed harmonised pheno- state of knowledge in diabetic neuropathy.
typing of neuropathy and neuropathic pain have
not yet been fully developed. As will be apparent
from other chapters in this book, the phenotype 2 A Brief Introduction
to Diabetes Genetics
A. Veluchamy · B. H. Smith
Division of Population Health and Genomics, School Partly for historical reasons there has been a
of Medicine, University of Dundee, dichotomy of approach in human genetics into,
Dundee, Scotland, UK firstly, the study of rare genetic disorders inher-
D. L. Bennett (*) ited in a monogenic (Mendelian) fashion, and
The Nuffield Department of Clinical Neuroscience, secondly, common disorders which are polygenic
University of Oxford, Oxford, UK with multiple genes each having a small impact
e-mail: david.bennett@ndcn.ox.ac.uk

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 239
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_14
240 A. Veluchamy et al.

on disease risk [1]. In Mendelian disorders the associated with type 1 diabetes and more than
disease allele is absent or extremely rare in the 200 with type 2 diabetes [8]. These are emphasis-
general population and has a large impact on ing important biological pathways such as pan-
gene function, for instance, through changes in creatic islet cell dysfunction in the case of type 2
the amino acid sequence or markedly reduced diabetes [9]. These advances are likely to con-
gene expression. A relevant example is neonatal tinue with increased application of genomics at
diabetes due to gain of function mutations in sub- population level (e.g., UK Biobank [10]) and
units of the ATP-sensitive potassium channel, a within national healthcare systems [11]. As we
critical determinant of excitability in pancreatic develop larger population cohorts, we are realis-
islet cells [5]. These mutations lead to reduced ing that, rather than a dichotomy of monogenic/
ATP sensitivity, reduced cellular excitability and Mendelian genetics versus polygenic/common
impaired insulin secretion. This finding provided disease genetics, there is a whole spectrum of
a molecular rationale for the use of Sulfonylurea effect sizes and allele frequencies within the pop-
drugs in this disorder, and is an excellent example ulation; gene variants may also have pleiotropic
of how gene identification can enlighten under- effects influencing multiple phenotypes/disor-
standing of disease pathophysiology and thera- ders. We are also rapidly seeing a transitioning of
peutics [6]. sequencing technologies (as costs drop) from
The second broad approach to human genetics array sequencing of SNPs and whole exome
is based on the fact that many common disorders sequencing to whole genome sequencing. Just as
are related to risk determined by multiple genes important as these changes in technologies are
(also interacting with environmental factors), and analysis techniques and availability of very large
this can be revealed by investigating the associa- datasets of genetic variation in adult populations
tion of the allele frequency of single nucleotide (gnomAD [12]) and in those with clinical disor-
polymorphisms (SNPs) in individuals with dis- ders (ClinVar [13]).
ease phenotype. Such associations can be studied
using a candidate gene approach or preferably
across the whole genome using a Genome-Wide 3 The Genomics of Diabetic
Association Study (GWAS) approach. Careful Neuropathy in Humans
attention to bias, appropriate significance thresh-
olds and large sample sizes have generated Genomic studies may help us to identify the
robust, reproducible findings across a range of genetic risk factors which result in 30–50% of
common diseases. In this context the SNPs iden- patients with diabetes developing neuropathy
tified each have a small effect size and in most [14]. This will help in our understanding of the
cases are non-coding, and their effect is thought pathogenesis of neuropathy, and any shared
to be mediated through subtle effects on gene genetic architecture with other diabetes compli-
expression through altering the function of gene cations, implying common pathophysiological
promoters and enhancers. Determining whether pathways.
and how a variant can impact on gene expression For the past two decades, many genetic asso-
is now greatly facilitated by large datasets being ciation studies have identified potential risk vari-
generated by consortia such as ENCODE, which ants for diabetic neuropathy. These indicate that
define the elements in DNA and histones that genetics has a significant contribution to its
regulate gene expression in multiple cell types development, though there are currently few
[7]. The GWAS approach requires large sample studies with heritability estimates. Table 1 pres-
sizes to generate sufficient statistical power. ents the known genetic variants associated with
Because diabetes is common and has a large neuropathy in people with type 2 diabetes, from
impact on health, diabetes has been at the fore- 25 human genetic association studies. The dia-
front of advances in genomics. More than 50 betic neuropathy phenotype was defined in most
genetic loci have been found to be significantly of the studies using at least one of the following:
The Genomics of Diabetic Neuropathy 241

Table 1  Known genetic variants associated with neuropathy in type 2 diabetes patients
Effect of
Study (year) Ethnicity N (cases/controls) Phenotyping Variants Gene name association
Arredondo- Mexican 218(90/128) Monofilament rs3025039 Vascular Protective
García et al. and tuning endothelial
(2019) [15] fork test growth factor
(VEGF)
Basol et al. Turkish 468(227/241) DNS and NDS VNTR Interleukin 4 Risk
(2013) [16] polymorphism (IL-4)
Buraczynska Polish 926(406/520) CNE and Pro198Leu Glutathione Risk
et al. (2017) EMG Peroxidase 1
[17] (GPx-1)
Chen et al. Chinese 160(80/80) WHO rs3821799 Adiponectin Risk
(2015) [18] diagnostic rs3774261 (ADPN)
criteria
Ciccacci et al. Italy 130(62/68) VPT and rs2910164 MIR146a Protective
(2014) [19] Neuropathic rs11888095 MIR128a Risk
questionnaire
Ciccacci et al. Italy 149(69/80) MNSI, VPT rs3746444 MIR499a Risk
(2018) [20]
Gupta and North 650(356/294) VPT and C106T Aldo-keto Risk
Singh (2017) Indian Monofilament rs759853 reductase
[21] family 1
member B
(AKR1B1)
Inanir et al. Turkish 516(235/281) NSS and NDS I/D Angiotensin- Risk
(2013) [22]a polymorphism converting
enzyme (ACE)
Ji et al. (2015) Chinese 180(90/90) Clinical T45G Adiponectin Risk
[23] assessment G276T (ADPN)
Kakavand Iranian 248(141/107) UKST C667T Methylene- Risk
Hamidi et al. score > 2, tetrahydrofolate
(2018) [24] microfilament reductase
(MTHFR)
Kolla et al. Indian 400(198/202) VPT +874A/T Interleukin 10 Risk
(2009) [25]a 1083G/A (IL-10)
Interferon-
gamma
(IFN-G)
Mansoor et al. Pakistan 772(276/496) Clinical I/D Angiotensin- Risk
(2012) [26] assessment converting
enzyme (ACE)
II
Marzban et al. Iranian 106(49/57) NSS and NDS HLA- Human Risk
(2016) [27] DQB1*02 and Leukocyte
HLA- Antigen (HLA)
DRB1*07
Monastiriotis Greece 234(54/180) NDS Epsilon 4 Apolipoprotein Risk
et al. (2013) E (APOE)
[28]
Papanas et al. Greek 130(70/60) DN index I/D Alpha2B Risk
(2007) [29] adrenergic
receptor
(ADRA2B)
Ren et al. Han 787(402/385) Monofilament rs5498 Intercellular Risk
(2015) [30] Chinese test, VPT, adhesion
Clinical molecule-1
assessment (ICAM-1)
(continued)
242 A. Veluchamy et al.

Table 1 (continued)
Effect of
Study (year) Ethnicity N (cases/controls) Phenotyping Variants Gene name association
Shah et al. Asian 495(139/356) VPT T-786C Endothelial- Risk
(2013) [31] Indian 475(133/342) (rs2070744), derived nitric
288(81/207) 27 VNTR oxide synthase
Intron 4, (eNOS)
G894T
(rs1799983)
Stephens et al. British 572(173/399) Clinical I/D Angiotensin- Risk
(2006) [32] assessment converting
enzyme (ACE)
Stoian et al. Spanish 174(84/90) Clinical I/D Vascular Risk
(2014) [33]a assessment Endothelial
Growth Factor
(VEGF)
Sun et al. Chinese 281(143/138) WHO rs2248069 Calcium Risk
(2018) [34] diagnostic rs16030 Voltage-Gated
Criteria rs216008 Channel
rs2239050 Subunit Alpha1
rs3794619 A (CACNA1A)
rs7191246 Calcium
Voltage-Gated
Channel
Subunit Alpha1
C (CACNA1C)
Calcium
Voltage-Gated
Channel
Subunit Alpha1
H (CACNA1H)
Tang et al. British 773(211/558) CNE rs1050450 Glutathione Risk
(2012) [35] peroxidase-1
(GPx-1)
Tang et al. American/ 5168(4384/784) MNSI >2.0 rs13417783 Sodium Risk
(2019) [36]b Canadian Voltage-Gated
Channel Alpha
Subunit 2
(SCN2A)
Ukinc et al. Turkish 52(37/15) Neurological C677T Methylene- Risk
(2009) [37] assessment and tetrahydrofolate
symptoms reductase
(MTHFR)
Wang et al. Chinese 251(101/150) Toronto C667T Methylene- Risk
(2012) [38] Diabetic tetrahydrofolate
Neuropathy reductase
Consensus (MTHFR)
Yigit et al. Turkish 512(230/282) NSS and NDS C667T Methylene- Risk
(2013) [22] tetrahydrofolate
reductase
(MTHFR)
ACCORD action to control cardiovascular risk in diabetes, BARI 2D bypass angioplasty revascularization investigation
in type 2 diabetes, CNE clinical neurological examination, DN diabetic neuropathy, CNE clinical neurological examina-
tion, EMG electromyography, MNSI Michigan neuropathy screening instrument, MPQ McGill pain questionnaire, NDS
neuropathy disability score, NSS neuropathy symptom score, UKST the United Kingdom screening test, VPT vibration
perception threshold, WHO World Health Organisation
a
DN vs healthy controls
b
Genome-wide association studies
The Genomics of Diabetic Neuropathy 243

monofilament test, vibration perception thresh- ined in four cohorts [22, 24, 37, 38]. The MTHFR
old, Toronto Diabetic Neuropathy Expert C677T polymorphism was found to be signifi-
Consensus, the United Kingdom screening test, cantly associated with diabetic neuropathy in a
Michigan neuropathy screening instrument meta-analysis of these cohorts (OR, 1.43;
(MNSI), neuropathy symptom score (NSS) and/ P = 0.014) [42]. Although the exact role of this
or neuropathy disability score (NDS). Most of gene in diabetic neuropathy is not yet known,
the candidate gene association studies investi- in vitro studies showed that hyper homocystein-
gated the association of genetic variants with dia- aemia affects nerve function.
betic neuropathy using cohorts of type 2 diabetes A recent meta-analysis [41], the GPx-1 vari-
patients with and without neuropathy. Very few ant (rs1050450) showed a significant association
compared diabetic neuropathy cases only with with diabetic neuropathy (OR = 1.43, P < 0.001).
healthy controls. The studies were conducted GPx-1 encodes an antioxidant enzyme that pro-
predominantly in cohorts of European and Asian tects cells against oxidative damage. Two other
ancestry. studies reported the association of two polymor-
Single nucleotide polymorphisms (SNPs) in phisms (I/D and rs3025039) in VEGF with dia-
angiotensin-converting enzyme (ACE), betic neuropathy [15, 33]. VEGF modulates
methylene-­tetrahydrofolate reductase (MTHFR), vascular permeability and contributes to neuro-
vascular endothelial growth factor (VEGF) and genesis. Further investigation is needed to vali-
glutathione peroxidase-1 (GPx-1) genes were date these genetic variants. Other reported genes
found to be associated with diabetic neuropathy associated with diabetic neuropathy in just a sin-
susceptibility in multiple studies of individuals gle study are presented in Table  1. ADRA2B,
from different ethnic backgrounds [39–41] (see CACNA1C, CACNA1H and CACNA1A are
Table  1). A recent systematic review and meta-­ mainly involved in neurotransmission. HLA-­
analysis reported that ACE I  >  D and MTHFR DQB1, HLA-DRB1-DQB1, IL-4, IFN-G, ICAM-­
1298A/C variants were significantly associated 1 and eNOS represent the immune response
with increased risk of diabetic neuropathy [42]. pathway. The other genes (APOE, ADPN and
The DD genotype of ACE was shown to increase AKR1B1) are relevant to cell signalling and
the risk of developing diabetic peripheral neu- metabolism. Notably, two studies implicated spe-
ropathy in Caucasian, South Asian, Turkish and cific microRNAs (rs2910164, rs11888095 and
Egyptian cohorts, with an odds ratio of 1.43  in rs3746444) involved in the regulation of inflam-
the meta-analysis (P  =  0.004) [42]. The ACE mation [19, 20]. None of these findings have yet
gene has been the most frequently studied in dia- been replicated in large cohorts.
betic neuropathy. It encodes angiotensin-­ Some candidate genes have been found to
converting enzyme which induces oxidative be associated with neuropathy in type 1 diabe-
stress, inflammation and vascular changes. ACE tes cohorts in Russia. These include the
inhibitors were shown to be effective in treating −262 T > C polymorphism of the catalase gene
diabetic neuropathy in experimental studies [43]. (CAT) [45, 46], the Ala(−9)Val SNP of super-
A recent study examined the efficacy of ACE oxide dismutase 2 (SOD) [47] and the
inhibitors for 2  years on 63 patients with auto- Arg213Gly polymorphism of superoxide dis-
nomic and/or peripheral diabetic neuropathy and mutase 3 (SOD3) [48, 49]. All three genes
found that they improved cardiovascular auto- (CAT, SOD2 and SOD3) protect the body
nomic diabetic neuropathy but did not improve against oxidative stress. A genetic variant
peripheral neuropathy [44]. More studies are (rs1001179) in CAT was shown to decrease the
therefore needed to elucidate the role of ACE risk of developing neuropathy in type 1 diabe-
inhibitors in diabetic neuropathy. The MTHFR tes (OR, 0.68; CI, 0.53–0.86; P  <  0.05) in a
gene, which encodes an enzyme that catalyses recent meta-analysis [41]. Two studies found
the conversion of homocysteine to methionine that the Arg allele of rs1799895 (Arg213Gly)
through the re-methylation pathway, was exam- in SOD3 increased the risk of developing neu-
244 A. Veluchamy et al.

ropathy in type 1 diabetes (OR, 5.91; P = 0.01 ing of diabetic neuropathy is hereditary sensory
[48] and OR, 1.64; P  =  0.02 [49]). Sample neuropathy type-1 (HSN1) a sub-type of CMT
sizes in these candidate gene association stud- with predominant sensory involvement. This is
ies ranged from 100 to 1000 and the findings caused by heterozygous mutations in the genes
are yet to be replicated. SPTLC1 [50, 51] or SPTLC2 [52] which encode
Recently, the first GWAS of diabetic neu- two of the three subunits which constitute the
ropathy found a novel genome-wide significant enzyme Serine Palmitoyl Transferase (SPT).
locus at chromosome 2q24 (P < 5 × 10−8) in the SPT has the highest affinity for l-serine and
Action to Control Cardiovascular Risk in palmitoyl-CoA forming formation of sphingoid
Diabetes (ACCORD) discovery cohort of 4384 bases (SBs) which are a class of lipid that form
white cases and 784 controls without neuropa- the backbone of all sphingolipids (SLs).
thy. The T allele of the lead SNP, r13417783, Interestingly HSN1 mutations alter the substrate
showed a protective effect with an odds ratio of specificity of SPT from the amino acid l-serine
0.64 (P = 1.9 × 10−9). The closest genes to the to l-alanine or l-glycine, resulting in the pro-
intergenic variant are Xin Actin Binding Repeat duction of the toxic deoxysphingoid bases
Containing 2 (XIRP2) and sodium voltage- (DSBs) [53]. The toxic DSBs can neither be
gated channel alpha subunit 2 (SCN2A) [36]. converted to complex sphingolipids nor be
The protective allele of this SNP influences the degraded, and hence they accumulate. HSN1
expression of SCN2A, which can increase neu- patients with SPTLC1 or 2 mutations have ele-
ronal excitability. It was replicated in an inde- vated plasma DSBs levels. There is a potential
pendent cohort of people with type 2 diabetes link between this metabolic pathway and dia-
and neuropathy (791 cases and 158 controls) in betic neuropathy because elevated plasma DSB
the Bypass Angioplasty Revascularization levels are also detected in the plasma of patients
Investigation in Type 2 Diabetes (BARI2D) with metabolic syndrome and type 2 diabetes
trial (OR, 0.57; P = 0.0009). It retained genome- [54, 55]. The DSB levels in patients with diabe-
wide significance (P = 7.9 × 10−12) in the meta- tes are not as high as those observed in HSN1
analysis (OR, 0.63). An MNSI score >2 was and as yet there is no direct causative link to dia-
used to define neuropathy cases in both cohorts. betic neuropathy. However these could be part
More GWAS are needed to validate this finding of the metabolic milieu which is injurious to
and identify more associated genetic variants. neurons in diabetes and understanding toxicity
mechanisms in HSN1 could be relevant to dia-
betic neuropathy.
3.1 Insights into Diabetic A further example of how understanding of
Neuropathy Provided by Gene CMT can inform our knowledge of diabetic neu-
Variants Associated ropathy is the recent finding that bi-allelic loss-­
with Charcot-Marie-Tooth of-­
function mutations in the sorbitol
dehydrogenase (SORD) gene is the most com-
Charcot-Marie-Tooth (CMT) is an umbrella mon cause of autosomal recessive inherited neu-
terms for inherited neuropathies and can poten- ropathy [56]. SORD is an enzyme that converts
tially inform our understanding of diabetic neu- sorbitol into fructose in the two-step polyol path-
ropathy. The most common form of CMT is a way which has previously been implicated in dia-
slowly progressive, length-dependent sensorim- betic neuropathy. These mutations lead to loss of
otor neuropathy with onset in the first two enzymatic activity, and participants with these
decades of life. There are sub-types with pre- mutations were found to have higher fasting
dominantly motor, sensory or autonomic pre- serum sorbitol levels. A Drosophila model could
sentations. More than 100 genes have now been recapitulate the phenotype which was rescued by
linked to CMT. An interesting example of how treatment with aldose reductase inhibitors (this
understanding CMT can inform our understand- drug class was originally developed for targeting
The Genomics of Diabetic Neuropathy 245

the polyol pathway in diabetic neuropathy). In rat transported down axons, potentially distorting
models of diabetes, inhibition of SORD worsens the results. This study also identified gene
neuropathy [57] and the toxicity of raised sorbitol expression patterns within nerves which were
is thought to be mediated by increased cellular predictive of neuropathy progression. Further
osmolarity and oxidative stress. Better under- studies are needed to see if such an approach can
standing of the mechanisms by which reduced be replicated in more accessible bio-samples
SORD levels result in CMT could therefore (such as skin biopsy or even gene expression
inform our understanding of diabetic profiling in blood).
neuropathy. The same sural nerve tissue was then used for
investigation of epigenetic factors by assessment
of DNA methylation [59]. DNA methylation
3.2 Insights Provided into refers to the covalent attachment of a methyl
Diabetic Neuropathy group to a cytosine adjacent to a guanine (CpG)
from Transcriptional Profiling by DNA methyltransferases. Methylation of
in Humans CpG islands (DNA regions with a high density
of CpG motifs) in the promoter of a gene
A further functional genomics approach to represses gene expression. A large number of
understand diabetic neuropathy is to assess differentially methylated CpGs were identified,
genome-­wide changes in gene expression evoked confirming epigenetic changes in the context of
by diabetic neuropathy. Changes in gene expres- progressive versus non-progressive diabetic neu-
sion reflect a complex mixture of genomic varia- ropathy. Functional analysis indicated that epi-
tion, epigenetic changes and the reaction to genes were enriched in biological pathways
metabolic changes/neural injury, but given the related to nervous system development, axon
strong link between gene expression and biolog- guidance, glycerophospholipid metabolism and
ical function helps provide important insight into MAPK signalling; several of which have previ-
disease pathogenesis. This approach is, however, ously been shown to be involved in diabetic
limited by access to human nerve tissue from peripheral neuropathy (DPN). A subsequent
subjects with diabetic neuropathy and relevant data-driven clustering approach was used and
controls. One study which met this challenge of found clusters defined by patterns of gene
tissue availability used tissue from participants expression which were related to high and low
who had taken part in a negative clinical trial levels of glycosylated haemoglobin (HbA1c)
which included two sural nerve biopsies, [60] which was then integrated with assessment
52  weeks apart. The authors compared gene of DNA methylation. There were 998 differen-
expression (assessed using micro-arrays) in the tially expressed and 929 differentially methyl-
sural nerves in those that had developed progres- ated genes when comparing the high and low
sive neuropathy versus those who had not, as HbA1c clusters. The immune system was identi-
defined by the change in myelinated fibre den- fied as an enriched pathway and the extracellular
sity [58]. Over 500 genes were found to be sig- matrix and axon guidance were also identified.
nificantly differentially expressed in the patient In summary, tissue availability has limited exam-
samples when comparing the progressive dia- ination of gene expression changes within the
betic neuropathy to non-progressive groups. nerve due to diabetic neuropathy, however, this
These were enriched for pathways involved in one cohort has been extensively studied and
the inflammatory response and lipid metabolism demonstrates extensive gene expression and epi-
and likely predominantly reflect gene expression genetic changes associated with neuropathy pro-
changes in Schwann cells, but it should be gression. These changes particularly highlight
remembered that these nerve biopsies will also alterations in the immune system/inflammation,
have contained fibroblasts, vascular and immune axon growth, extracellular matrix and lipid
cells; furthermore, some mRNAs are actively metabolism.
246 A. Veluchamy et al.

4 The Genomics These signals may be specific to neuropathic


of Neuropathic Pain pain and warrant further studies.
in Diabetic Neuropathy A candidate gene association study by Ursu
in Humans et al. found an association between the presence
of the gain of function SNP, rs1718119
Neuropathic pain was shown to have a heritabil- (Ala348Thr), in the purinergic receptor 7 gene
ity of 37% in a twins study [61]. In recent years, (P2RX7) and higher pain intensity score in
genetic studies have identified a number of females with diabetic neuropathy (P  =  0.039)
genetic polymorphisms associated with various [65]. In this study, 156 patients with diabetic neu-
common neuropathic pain conditions but few of ropathic pain were identified by their MNSI score
these studies have examined neuropathic pain in and 24-h average pain severity rating. One of the
diabetic populations [62]. These genes are asso- most studied variants in other chronic pain condi-
ciated with ion channels, and/or are involved tions, rs1799971, A118G in the Mu-opioid recep-
with neurotransmission, metabolic and immune tor gene, was shown to be associated with
response pathways. There is a general lack of neuropathic pain in patients with a diabetic foot
replication of these findings, again through lack ulcer (OR, 0.24; P = 0.038) [66]. However, each
of harmonised phenotyping and sample sizes of these variants has only been identified by a
with sufficient statistical power. single study, each with a small sample size. More
Meng et al. reported the first GWAS of neu- studies are required to find reliable genetic varia-
ropathic pain in patients with diabetic neuropa- tions for diabetic neuropathic pain. Harnessing
thy (572 cases and 2491 controls) phenotyped modern genomic technologies and aggregating
using medication and monofilament test records. large cohorts with a validated phenotype can pro-
They found a suggestive variant at chromosome vide better understanding of the genetic architec-
8p21.3 (rs17428041; OR, 0.67; P  =  1  ×  10−7) ture of neuropathic pain in diabetic neuropathy.
near the glial cell-derived neurotrophic factor
family receptor alpha 2 gene (GFRA2), which is
involved in receptor signalling and the immune 4.1 Sodium Channel Variants
response [63]. In the same diabetes cohort, a and Painful Diabetic
gender-­ specific GWAS reported a SNP-based Neuropathy
heritability of 30% and 14.7% in males and
females respectively. This study identified a Voltage-gated sodium channels are key determi-
suggestive SNP at chromosome 8q23.1 nants of sensory neuron excitability and are
(rs6986153; OR, 1.67; P = 8.02 × 10−7) near the required for the initial amplification of the gen-
high-mobility group box 1 pseudogene 46 erator potential within nociceptor terminals, the
(HMGB1P46) in males (470 cases and 2021 production of regenerative action potentials to
controls) and at chromosome 1q35.1 transmit information along axons to the CNS,
(rs71647933; OR, 2.31; P  =  2.75  ×  10−7) near and ultimately the ingress of action potentials
the zinc-finger and SCAN domain-encoding- into the central terminals and neurotransmitter
toll-like receptor 12 pseudogene (ZSCAN20- release [67]. The voltage-gated sodium channel
TLR12P) in females (491 cases and 1239 consists of a pore-forming α-subunit complexed
controls) [63]. These genes are involved in the to two non-pore-forming β-subunits (which
immune response pathway. Both studies lack modulate trafficking and function of the
genome-wide signals for neuropathic pain sus- α-subunit). There are nine genes encoding the
ceptibility and failed to replicate in other neuro- α-subunits (Nav1.1–1.9) and a number of these
pathic pain conditions [64]. Moreover, these (Nav1.7, 1.8 and 1.9) are highly and relatively
suggestive signals were not shown to be associ- selectively expressed within nociceptors. Human
ated with diabetic neuropathy in a more recent genetics has demonstrated a strong association
GWAS of diabetic peripheral neuropathy [36]. between variants in these genes and pain percep-
The Genomics of Diabetic Neuropathy 247

tion. Bi-allelic loss-of-function mutations in been linked to painful neuropathy (M1852T and
Nav1.7 results in congenital insensitivity to pain T1596I), which demonstrated markedly impaired
[68] whilst gain of function variants in Nav1.7 channel fast inactivation on patch clamp analysis
have been linked with the pain disorders erythro- resulting in gain of function. This is likely to be a
melalgia (painful, red extremities exacerbated gene-environment interaction in that participants
by warmth [69]), paroxysmal extreme pain dis- did not report pain until they developed diabetic
order (paroxysmal, pain and erythema of the neuropathy. Thus, although the Nav1.7 variants
mandibular, ocular and perineal regions often resulted in hyper-excitability this was tolerated in
triggered by mechanical stimulation [70]) and early life, but the added ‘stress’ of diabetes
small fibre neuropathy (pain associated with appeared to trigger a chronic pain state. Not all
degeneration of small fibre terminals in the skin studies have found an association between rare
[71]). Gain of function variants in Nav1.8 and variants in voltage-gated sodium channels
Nav1.9 have also been linked to painful neuropa- (VGSCs) and painful neuropathy. Wadgawan
thies [72, 73]. et al., reported no difference in the frequency of
Given this human genetic data linking sodium rare variants in Nav1.7, 1.8 and 1.9  in painful
channel variants to pain, it is a natural question as versus painless neuropathy of mixed aetiology
to whether variants in sodium channels alter risk [76]. This total cohort of 457 included 179 with
or severity of neuropathic pain in the context of diabetic neuropathy, but the diabetic neuropathy
diabetic neuropathy. A number of studies have group was not analysed independently. Further
investigated this issue. Li et  al., compared a studies on larger well-phenotyped cohorts are
cohort of almost 1000 participants with painful needed.
diabetic neuropathy that had been recruited in tri- Genetic variation in VGSCs relevant to neuro-
als to over 3000 population controls [74] and pathic pain may not be restricted to α-subunits.
found a higher frequency of the Nav1.7 variants Recently a variant (D109N) in the β2-subunit
Val991Leu/Met932Leu in cases versus controls; was identified in a participant with painful dia-
these variants were also associated with more betic neuropathy [77]. The β-subunits are not
severe neuropathic pain. These variants are in pore-forming but complex with and sculpt the
complete linkage disequilibrium (i.e., always functional properties of pore-forming α-subunits.
inherited together). Interestingly Val991Leu/ This variant was found to result in hyper-­
Met932Leu had previously been identified in excitability when expressed in DRG neurons due
patients with idiopathic small fibre neuropathy to a gain of function effect on Nav1.7.
and shown to produce gain of function in terms of Identification of variants in VGSCs may have
enhancing resurgent currents and causing hyper-­ implications for treatment of neuropathic pain.
excitability when expressed in rat dorsal root Idiopathic small fibre neuropathy has already
ganglion (DRG) neurons [71]. A further study provided an example of a stratified pain medi-
used targeted next generation sequencing to cine approach in which lacosamide, which acts
examine rare variants (present in less than 0.5% as a blocker of VGSCs but has limited efficacy
of the general population) in a cohort of partici- in unselected neuropathic pain patients, was
pants with diabetic neuropathy in which detailed shown to be effective versus placebo in a
phenotyping for neuropathic pain had been per- selected group of patients with painful small
formed [75]. Rare variants in Nav1.7 were more fibre neuropathy associated with rare variants in
common in the painful versus painless diabetic Nav1.7 [78]. In certain rare cases personalised
neuropathy group. A number of the variants iden- treatment approaches may be taken: for exam-
tified in the painful diabetic neuropathy group ple, an S242T variant in Nav1.8 was identified
had previously been identified in painful idio- in a participant with painful diabetic neuropathy
pathic small fibre neuropathy and found to pro- [79]. Interestingly the equivalent amino acid
duce gain of function. A number of novel variants change (S241T) in Nav1.7 had previously been
were also identified, which had not previously linked to inherited erythromelalgia and found to
248 A. Veluchamy et al.

cause gain of function which was responsive to Acknowledgements This work was supported by the
European Union’s Horizon 2020 research and innovation
carbamazepine (which has activity against programme under grant agreement No 633491
VGSCs but is not routinely used in the treatment (DOLORisk).
of painful diabetic neuropathy). Structural mod-
elling comparing S242T Nav1.8 to S241T
Nav1.7 showed that these were indeed equiva- References
lent amino acids in these two ion channels, in
the same orientation and electrophysiological 1. Claussnitzer M, Cho JH, Collins R, Cox NJ,
analysis demonstrated that S242T Nav1.8 also Dermitzakis ET, Hurles ME, Kathiresan S, Kenny EE,
Lindgren CM, MacArthur DG, et al. A brief history
resulted in gain of function which could be of human disease genetics. Nature. 2020;577:179–89.
reversed by carbamazepine. 2. Ahlqvist E, van Zuydam NR, Groop LC, McCarthy
MI. The genetics of diabetic complications. Nat Rev
Nephrol. 2015;11:277–87.
3. Finnerup NB, Haroutounian S, Kamerman P, Baron
5 Conclusion R, Bennett DL, Bouhassira D, Cruccu G, Freeman R,
Hansson P, Nurmikko T, et  al. Neuropathic pain: an
We are in the early phase of applying genomic updated grading system for research and clinical prac-
technologies to diabetic neuropathy and the asso- tice. Pain. 2016;157:1599–606.
4. van Hecke O, Kamerman PR, Attal N, Baron R,
ciated complication of neuropathic pain. Large Bjornsdottir G, Bennett DL, Bennett MI, Bouhassira
cohorts with detailed harmonised phenotyping of D, Diatchenko L, Freeman R, et al. Neuropathic pain
neuropathy and neuropathic pain have only phenotyping by international consensus (NeuroPPIC)
recently been developed and so we are now likely for genetic studies: a NeuPSIG systematic review,
Delphi survey, and expert panel recommendations.
to see an accelerating pace of discovery. There Pain. 2015;156:2337–53.
have been some early encouraging findings, for 5. Gloyn AL, Pearson ER, Antcliff JF, Proks P, Bruining
instance, the GWAS of diabetic neuropathy iden- GJ, Slingerland AS, Howard N, Srinivasan S, Silva
tifying a SNP which is in proximity to and alters JM, Molnes J, et al. Activating mutations in the gene
encoding the ATP-sensitive potassium-channel sub-
the expression of SCN2A (encoding Nav1.2). unit Kir6.2 and permanent neonatal diabetes. N Engl
Rare variants in voltage-gated sodium channels J Med. 2004;350:1838–49.
(especially Nav1.7) have also been linked to 6. Pipatpolkai T, Usher S, Stansfeld PJ, Ashcroft
painful (versus painless) diabetic neuropathy in FM. New insights into KATP channel gene mutations
and neonatal diabetes mellitus. Nat Rev Endocrinol.
candidate gene studies. These lead to gain of 2020;16:378–93.
channel function so providing a plausible func- 7. Consortium EP.  An integrated encyclopedia of
tional link to sensory neuron hyper-excitability DNA elements in the human genome. Nature.
and pain. We have also learned lessons as to how 2012;489:57–74.
8. Mahajan A, Taliun D, Thurner M, Robertson NR,
metabolism can have a major impact on nerve Torres JM, Rayner NW, Payne AJ, Steinthorsdottir V,
function through understanding the genetic basis Scott RA, Grarup N, et al. Fine-mapping type 2 diabe-
of neuropathies inherited in a Mendelian fashion tes loci to single-variant resolution using high-density
(such as CMT) and interestingly parallels are imputation and islet-specific epigenome maps. Nat
Genet. 2018;50:1505–13.
arising with some of these metabolic changes to 9. Krentz NAJ, Gloyn AL. Insights into pancreatic islet
those observed in diabetes. As this field matures cell dysfunction from type 2 diabetes mellitus genet-
advances in genomics could have significant ics. Nat Rev Endocrinol. 2020;16:202–12.
impact in the clinical management of diabetic 10. Sudlow C, Gallacher J, Allen N, Beral V, Burton P,
Danesh J, Downey P, Elliott P, Green J, Landray M,
neuropathy and pain: including improved under- et al. UK biobank: an open access resource for identi-
standing of the pathogenesis of these disorders fying the causes of a wide range of complex diseases
and discovery of novel treatment targets, the of middle and old age. PLoS Med. 2015;12:e1001779.
identification of those most at risk of neuropathy 11. Turro E, Astle WJ, Megy K, Graf S, Greene D,
Shamardina O, Allen HL, Sanchis-Juan A, Frontini
and pain in order to intervene at an early stage M, Thys C, et  al. Whole-genome sequencing of
and a more personalised approach to treatment patients with rare diseases in a national health system.
selection. Nature. 2020;583:96–102.
The Genomics of Diabetic Neuropathy 249

12. Karczewski KJ, Francioli LC, Tiao G, Cummings 23. Ji ZY, Li HF, Lei Y, Rao YW, Tan ZX, Liu HJ, Yao
BB, Alfoldi J, Wang Q, Collins RL, Laricchia KM, GD, Hou B, Sun ML. Association of adiponectin
Ganna A, Birnbaum DP, et  al. The mutational con- gene polymorphisms with an elevated risk of diabetic
straint spectrum quantified from variation in 141,456 peripheral neuropathy in type 2 diabetes patients. J
humans. Nature. 2020;581:434–43. Diabetes Complications. 2015;29(7):887–92. https://
13. Landrum MJ, Chitipiralla S, Brown GR, Chen C, Gu doi.org/10.1016/j.jdiacomp.2015.06.008. Epub 2015
B, Hart J, Hoffman D, Jang W, Kaur K, Liu C, et al. Jun 19. PMID:26144281.
ClinVar: improvements to accessing data. Nucleic 24. Kakavand Hamidi A, Radfar M, Amoli
Acids Res. 2020;48:D835–44. MM.  Association between MTHFR variant and dia-
14. Feldman EL, Callaghan BC, Pop-Busui R, Zochodne betic neuropathy. Pharmacol Rep. 2018;70:1–5.
DW, Wright DE, Bennett DL, Bril V, Russell JW, 25. Kolla VK, Madhavi G, Pulla Reddy B, Srikanth Babu
Viswanathan V.  Diabetic neuropathy. Nat Rev Dis BM, Yashovanthi J, Valluri VL, Ramesh J, Akka J.
Primers. 2019;5:41. Association of tumor necrosis factor alpha, interferon
15. Arredondo-García VK, Cepeda-Nieto AC, Batallar-­ gamma and interleukin 10 gene polymorphisms with
Gómez T, Salinas-Santander M, Zugasti-Cruz A, peripheralneuropathy in South Indian patients with
Ramírez-Calvillo L, Maldonado-Sánchez K, Morlett-­ type 2 diabetes. Cytokine. 2009;47(3):173–7. https://
Chávez J, Barajas-Martínez H.  Association of the doi.org/10.1016/j.cyto.2009.06.007. Epub 2009 Jul
vascular endothelial growth factor gene polymor- 15. PMID: 19608431.
phism +936 C/T with diabetic neuropathy in Mexican 26. Mansoor Q, Javaid A, Bilal N, Ismail M. Angiotensin-
patients with Type 2 diabetes mellitus. Arch Med Res. converting enzyme (ACE) gene II genotype pro-
2019;50:181–6. tects against the development of diabetic peripheral
16. Basol N, Inanir A, Yigit S, Karakus N, Kaya SU. High neuropathy in type 2 diabetes mellitus. J Diabetes.
association of IL-4 gene intron 3 VNTR polymorphism 2012;4(3):257–61. https://doi.org/10.1111/j.1753-
with diabetic peripheral neuropathy. J Mol Neurosci. 0407.2012.00205.x. PMID: 22607040.
2013;51(2):437–41. https://doi.org/10.1007/s12031- 27. Marzban A, Kiani J, Hajilooi M, Rezaei H,
013-0048-y. Epub 2013 Jun 16. PMID: 23771787. Kahramfar Z, Solgi G. HLA class II alleles and
17. Buraczynska M, Buraczynska K, Dragan M, Ksiazek risk for peripheral neuropathy in type 2 diabetes
A. Pro198Leu Polymorphism in the glutathione per- patients. Neural Regen Res. 2016;11(11):1839–44.
oxidase 1 gene contributes to diabetic peripheral neu- https://doi.org/10.4103/1673-5374.194756. PMID:
ropathy in type 2 diabetes patients. Neuromolecular 28123430.
Med. 2017;19(1):147–53. https://doi.org/10.1007/ 28. Monastiriotis C, Papanas N, Trypsianis G,
s12017-016-8438-2. Epub 2016 Sep 3. PMID: Karanikola K, Veletza S, Maltezos E. The ε4
27592002. allele of the APOE gene is associated with more
18. Chen Y, Wang J, Wang L, Huang P, Tan ZX, Liu HJ. severe peripheral neuropathy in type 2 diabetic
Adiponectin gene polymorphisms are associated with patients. Angiology. 2013;64(6):451–5. https://doi.
increased susceptibility to diabetic peripheral neu- org/10.1177/0003319712453645. Epub 2012 Jul 23.
ropathy. Biomarkers. 2015;20(6–7):474–80. https:// PMID: 22826377.
doi.org/10.3109/1354750X.2015.1101784. PMID: 29. Papanas N, Papatheodorou K, Papazoglou D,
26616148. Kotsiou S, Christakidis D, Maltezos E. An insertion/
19. Ciccacci C, Morganti R, Di Fusco D, D'Amato C, deletion polymorphism in the alpha2B adrenocep-
Cacciotti L, Greco C, Rufini S, Novelli G, Sangiuolo tor gene is associated with peripheral neuropathy
F, Marfia GA, et  al. Common polymorphisms in in patients with type 2 diabetes mellitus. Exp Clin
MIR146a, MIR128a and MIR27a genes contribute Endocrinol Diabetes. 2007;115(5):327–30. https://
to neuropathy susceptibility in type 2 diabetes. Acta doi.org/10.1055/s-2007-967084. PMID:17516297.
Diabetol. 2014;51:663–71. 30. Ren Z, Ji N, Jia K, et al. Association of the intercel-
20. Ciccacci C, Latini A, Greco C, Politi C, D’Amato lular adhesion molecule-1 gene polymorphisms with
C, Lauro D, Novelli G, Borgiani P, Spallone type 2 diabetes and diabetic peripheral neuropa-
V.  Association between a MIR499A polymorphism thy in a Chinese Han population. Genes Genomics.
and diabetic neuropathy in type 2 diabetes. J Diabetes 2015;37:69–75.
Complicat. 2018;32(1):11–7. 31. Shah VN, Cheema BS, Kohli HS, Sharma R, Khullar
21. Gupta B, Singh SK. Association of aldose reductase M, Bhansali A. Endothelial nitric oxide synthase gene
gene polymorphism (C-106T) in susceptibility of dia- polymorphism and the risk of diabetic neuropathy in
betic peripheral neuropathy among north Indian popu- Asian Indian patients with type 2 diabetes. J Diabetes
lation. J Diabetes Complications. 2017;31(7):1085–9. Metab. 2013;4:243.
https://doi.org/10.1016/j.jdiacomp.2017.04.011. 32. Stephens JW, Dhamrait SS, Acharya J, Humphries
Epub 2017 Apr 28. PMID: 28495421. SE, Hurel SJ. A common variant in the ACE gene is
22. Yigit S, Karakus N, Inanir A.  Association of associated with peripheral neuropathy in women with
MTHFR gene C677T mutation with diabetic periph- type 2 diabetes mellitus. J Diabetes Complications.
eral neuropathy and diabetic retinopathy. Mol Vis. 2006;20(5):317–21. https://doi.org/10.1016/j.jdia-
2013;19:1626–30. comp.2005.07.010. PMID: 16949519.
250 A. Veluchamy et al.

33. Stoian A, Bacârea A, Moţǎţǎianu A, Stoian M, Gliga Mourouglakis A, Hatzitolios AI. Efficacy of adminis-
F, Bacârea V, Carmen D, Bǎnescu C. Vascular endo- tration of an angiotensin converting enzyme inhibitor
thelial growth factor insertion/deletion gene polymor- for two years on autonomic and peripheral neuropa-
phism in patients with type 2 diabetes and diabetic thy in patients with diabetes mellitus. J Diabetes Res.
peripheral polyneuropathy. Rev Romana Med Lab. 2017;2017:6719239.
2014;22:165–72. 45. Babizhayev MA, Strokov IA, Nosikov VV, Savel’yeva
34. Sun L, Ma J, Mao Q, Yang YL, Ma LL, Niu L, Liu LF. EL, Sitnikov VF, Yegorov YE, Lankin VZ. The role
Association of single nucleotide polymorphisms in of oxidative stress in diabetic neuropathy: generation
CACNA 1A/CACNA 1C/CACNA 1H calcium channel of free radical species in the glycation reaction and
genes with diabetic peripheral neuropathy in Chinese gene polymorphisms encoding antioxidant enzymes
population. Biosci Rep. 2018;38(3):BSR20171670. to genetic susceptibility to diabetic neuropathy in
https://doi.org/10.1042/BSR20171670. Print 2018 population of type I diabetic patients. Cell Biochem
Jun 29. PMID: 29581247. Biophys. 2015;71(3):1425–43.
35. Tang TS, Prior SL, Li KW, Ireland HA, Bain SC, 46. Kasznicki J, Sliwinska A, Kosmalski M, Merecz A,
Hurel SJ, Cooper JA, Humphries SE, Stephens JW. Majsterek I, Drzewoski J. Genetic polymorphisms
Association between the rs1050450 glutathione per- (Pro197Leu of Gpx1, +35A/C of SOD1, -262C/T of
oxidase-1 (C > T) gene variant and peripheral neu- CAT), the level of antioxidant proteins (GPx1, SOD1,
ropathy in two independent samples of subjects CAT) and the risk of distal symmetric polyneuropathy
with diabetes mellitus. Nutr Metab Cardiovasc Dis. in Polish patients with type 2 diabetes mellitus. Adv
2012;22(5):417–25. https://doi.org/10.1016/j. Med Sci. 2016;61(1):123–9.
numecd.2010.08.001. Epub 2010 Dec 24. PMID: 47. Chistyakov DA, Savost’anov KV, Zotova EV, Nosikov
21185702. VV.  Polymorphisms in the Mn-SOD and EC-SOD
36. Tang Y, Lenzini PA, Pop-Busui R, Ray PR, Campbell genes and their relationship to diabetic neuropathy in
H, Perkins BA, Callaghan B, Wagner MJ, Motsinger-­ type 1 diabetes mellitus. BMC Med Genet. 2001;2:4.
Reif AA, Buse JB, et al. A genetic locus on chromo- 48. Strokov IA, Bursa TR, Drepa OI, Zotova VE, Nosikov
some 2q24 predicting peripheral neuropathy risk in VV, Ametov AS. Predisposing genetic factors for dia-
type 2 diabetes: results from the ACCORD and BARI betic polyneuropathy in patients with type 1 diabetes:
2D studies. Diabetes. 2019;68:1649–62. a population-based case-control study. Acta Diabetol.
37. Ukinc K, Ersoz HO, Karahan C, Erem C, 2003;40:375–9.
Eminagaoglu S, Hacihasanoglu AB, Yilmaz M, 49. Zotova VE, Chistyakov DA, Savost'yanov VK, Bursa
Kocak M.  Methyltetrahydrofolate reductase C677T TR, Galeev VI, Strokov IA, Nosikov VV. Association
gene mutation and hyperhomocysteinemia as a novel of the SOD2 Ala(−9)Val and SOD3 Arg213Gly poly-
risk factor for diabetic nephropathy. Endocrine. morphisms with diabetic polyneuropathy in diabetes
2009;36:255–61. mellitus type 1. Mol Biol. 2003;37:404–8.
38. Wang H, Fan D, Hong T.  Is the C677T polymor- 50. Bejaoui K, Wu C, Scheffler MD, Haan G, Ashby P,
phism in methylenetetrahydrofolate reductase gene or Wu L, de Jong P, Brown RH Jr. SPTLC1 is mutated
plasma homocysteine a risk factor for diabetic periph- in hereditary sensory neuropathy, type 1. Nat Genet.
eral neuropathy in Chinese individuals? Neural Regen 2001;27:261–2.
Res. 2012;7:2384–91. 51. Dawkins JL, Hulme DJ, Brahmbhatt SB, Auer-­
39. Politi C, Ciccacci C, D'Amato C, Novelli G, Borgiani Grumbach M, Nicholson GA. Mutations in SPTLC1,
P, Spallone V.  Recent advances in exploring the encoding serine palmitoyltransferase, long chain base
genetic susceptibility to diabetic neuropathy. Diabetes subunit-1, cause hereditary sensory neuropathy type
Res Clin Pract. 2016;120:198–208. I. Nat Genet. 2001;27:309–12.
40. Witzel II, Jelinek HF, Khalaf K, Lee S, Khandoker 52. Rotthier A, Auer-Grumbach M, Janssens K, Baets J,
AH, Alsafar H.  Identifying common genetic risk Penno A, Almeida-Souza L, Van Hoof K, Jacobs A,
factors of diabetic neuropathies. Front Endocrinol. De Vriendt E, Schlotter-Weigel B, et al. Mutations in
2015;6:1–18. the SPTLC2 subunit of serine palmitoyltransferase
41. Zhao Y, Zhu R, Wang D, Liu X. Genetics of diabetic cause hereditary sensory and autonomic neuropathy
neuropathy: systematic review, meta-analysis and type I. Am J Hum Genet. 2010;87:513–22.
trial sequential analysis. Ann Clin Transl Neurol. 53. Penno A, Reilly MM, Houlden H, Laura M, Rentsch
2019;6:1996–2013. K, Niederkofler V, Stoeckli ET, Nicholson G, Eichler
42. Wu S, Han Y, Hu Q, Zhang X, Cui G, Li Z, Guan F, Brown RH Jr, et al. Hereditary sensory neuropathy
Y. Effects of common polymorphisms in the MTHFR type 1 is caused by the accumulation of two neurotoxic
and ACE genes on diabetic peripheral neuropa- sphingolipids. J Biol Chem. 2010;285:11178–87.
thy progression: a meta-analysis. Mol Neurobiol. 54. Bertea M, Rutti MF, Othman A, Marti-Jaun J,
2017;54:2435–44. Hersberger M, von Eckardstein A, Hornemann
43. James E, Vincent A, Cheng T, Feldman EL. Diabetic T. Deoxysphingoid bases as plasma markers in diabe-
neuropathy: mechanisms to management. Pharmacol tes mellitus. Lipids Health Dis. 2010;9:84.
Ther. 2008;120:1–34. 55. Dohrn MF, Othman A, Hirshman SK, Bode H,
44. Didangelos T, Tziomalos K, Margaritidis C, Alecu I, Fahndrich E, Karges W, Weis J, Schulz JB,
Kontoninas Z, Stergiou I, Tsotoulidis S, Karlafti E, Hornemann T, et  al. Elevation of plasma 1-deoxy-­
The Genomics of Diabetic Neuropathy 251

sphingolipids in type 2 diabetes mellitus: a suscepti- 68. Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts
bility to neuropathy? Eur J Neurol. 2015;22:806–14, E, Springell K, Karbani G, Jafri H, Mannan J, Raashid
e855 Y, et al. An SCN9A channelopathy causes congenital
56. Cortese A, Zhu Y, Rebelo AP, Negri S, Courel S, Abreu inability to experience pain. Nature. 2006;444:894–8.
L, Bacon CJ, Bai Y, Bis-Brewer DM, Bugiardini E, 69. Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan J, Bu
et al. Biallelic mutations in SORD cause a common D, Liu B, Fan Z, et al. Mutations in SCN9A, encod-
and potentially treatable hereditary neuropathy with ing a sodium channel alpha subunit, in patients with
implications for diabetes. Nat Genet. 2020;52:473–81. primary erythermalgia. J Med Genet. 2004;41:171–4.
57. Schmidt RE, Dorsey DA, Beaudet LN, Plurad SB, 70. Fertleman CR, Baker MD, Parker KA, Moffatt S,
Parvin CA, Yarasheski KE, Smith SR, Lang HJ, Elmslie FV, Abrahamsen B, Ostman J, Klugbauer N,
Williamson JR, Ido Y. Inhibition of sorbitol dehydro- Wood JN, Gardiner RM, et al. SCN9A mutations in
genase exacerbates autonomic neuropathy in rats with paroxysmal extreme pain disorder: allelic variants
streptozotocin-induced diabetes. J Neuropathol Exp underlie distinct channel defects and phenotypes.
Neurol. 2001;60:1153–69. Neuron. 2006;52:767–74.
58. Hur J, Sullivan KA, Pande M, Hong Y, Sima AA, 71. Faber CG, Hoeijmakers JG, Ahn HS, Cheng X,
Jagadish HV, Kretzler M, Feldman EL.  The identi- Han C, Choi JS, Estacion M, Lauria G, Vanhoutte
fication of gene expression profiles associated with EK, Gerrits MM, et  al. Gain of function Nanu1.7
progression of human diabetic neuropathy. Brain. mutations in idiopathic small fiber neuropathy. Ann
2011;134:3222–35. Neurol. 2012a;71:26–39.
59. Guo K, Elzinga S, Eid S, Figueroa-Romero C, Hinder 72. Faber CG, Lauria G, Merkies IS, Cheng X, Han C,
LM, Pacut C, Feldman EL, Hur J.  Genome-wide Ahn HS, Persson AK, Hoeijmakers JG, Gerrits MM,
DNA methylation profiling of human diabetic periph- Pierro T, et  al. Gain-of-function Nav1.8 mutations
eral neuropathy in subjects with type 2 diabetes mel- in painful neuropathy. Proc Natl Acad Sci U S A.
litus. Epigenetics. 2019;14:766–79. 2012b;109:19444–9.
60. Guo K, Eid SA, Elzinga SE, Pacut C, Feldman EL, 73. Huang J, Han C, Estacion M, Vasylyev D, Hoeijmakers
Hur J.  Genome-wide profiling of DNA methyla- JG, Gerrits MM, Tyrrell L, Lauria G, Faber CG, Dib-­
tion and gene expression identifies candidate genes Hajj SD, et al. Gain-of-function mutations in sodium
for human diabetic neuropathy. Clin Epigenetics. channel Na(v)1.9  in painful neuropathy. Brain.
2020;12:123. 2014;137:1627–42.
61. Momi SK, Fabiane SM, Lachance G, Livshits G, 74. Li QS, Cheng P, Favis R, Wickenden A, Romano
Williams FM.  Neuropathic pain as part of chronic G, Wang H.  SCN9A variants may be implicated in
widespread pain: environmental and genetic influ- neuropathic pain associated with diabetic periph-
ences. Pain. 2015;156:2100–6. eral neuropathy and pain severity. Clin J Pain.
62. Veluchamy A, Hebert HL, Meng W, Palmer CNA, 2015;31:976–82.
Smith BH.  Systematic review and meta-analysis 75. Blesneac I, Themistocleous AC, Fratter C, Conrad
of genetic risk factors for neuropathic pain. Pain. LJ, Ramirez JD, Cox JJ, Tesfaye S, Shillo PR, Rice
2018;159:825–48. ASC, Tucker SJ, et al. Rare NaV1.7 variants associ-
63. Meng W, Deshmukh H, Donnelly LA, Torrance N, ated with painful diabetic peripheral neuropathy. Pain.
Colhoun HM, Palmer CNA, Smith BH.  A genome-­ 2018;159:469–80.
wide association study provides evidence of 76. Wadhawan S, Pant S, Golhar R, Kirov S, Thompson
sex-­specific involvement of Chr1p35.1 (ZSCAN20-­ J, Jacobsen L, Qureshi I, Ajroud-Driss S, Freeman R,
TLR12P) and Chr8p23.1 (HMGB1P46) with diabetic Simpson DM, et al. NaV channel variants in patients
neuropathic pain. EBioMedicine. 2015;2:1386–93. with painful and nonpainful peripheral neuropathy.
64. Warner SC, van Meurs JB, Schiphof D, Bierma-­ Neurol Genet. 2017;3:e207.
Zeinstra SM, Hofman A, Uitterlinden AG, Richardson 77. Alsaloum M, Estacion M, Almomani R, Gerrits MM,
H, Jenkins W, Doherty M, Valdes AM. Genome-wide Bonhof GJ, Ziegler D, Malik R, Ferdousi M, Lauria
association scan of neuropathic pain symptoms post G, Merkies IS, et al. A gain-of-function sodium chan-
total joint replacement highlights a variant in the nel beta2-subunit mutation in painful diabetic neurop-
protein-­kinase C gene. Eur J Hum Genet. 2017;44:1–6. athy. Mol Pain. 2019;15:1744806919849802.
65. Ursu D, Ebert P, Langron E, Ruble C, Munsie L, Zou 78. de Greef BTA, Hoeijmakers JGJ, Geerts M, Oakes M,
W, Fijal B, Qian Y-W, McNearney TA, Mogg A, et al. Church TJE, Waxman SG, Dib-Hajj SD, Faber CG,
Gain and loss of function of P2X7 receptors: mecha- Merkies ISJ.  Lacosamide in patients with Nav1.7
nisms, pharmacology and relevance to diabetic neuro- mutations-related small fibre neuropathy: a random-
pathic pain. Mol Pain. 2014;10:37. ized controlled trial. Brain. 2019;142:263–75.
66. Cheng KI, Lin SR, Chang LL, Wang JY, Lai 79. Han C, Themistocleous AC, Estacion M, Dib-Hajj
CS.  Association of the functional A118G polymor- FB, Blesneac I, Macala L, Fratter C, Bennett DL,
phism of OPRM1 in diabetic patients with foot ulcer Waxman SG, Dib-Hajj SD.  The novel activity of
pain. J Diabetes Complicat. 2010;24:102–8. carbamazepine as an activation modulator extends
67. Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-­ from NaV1.7 mutations to the NaV1.8-S242T mutant
Hajj SD. The role of voltage-gated sodium channels channel from a patient with painful diabetic neuropa-
in pain signaling. Physiol Rev. 2019;99:1079–151. thy. Mol Pharmacol. 2018;94:1256–69.
Metabolic Mechanisms in Diabetic
Neuropathy

Mark Yorek

1 Introduction dependent neuropathy [4, 5]. This decrease in


sensory perception is the most common and ear-
Diabetic peripheral neuropathy is a heterogenous liest form of diabetic peripheral neuropathy and
condition that can manifest as many different is gradual with symptoms of tingling, pain, and
symptoms and are the most common complica- loss of sensation in the toes [6, 7]. Clinical evi-
tions of diabetes mellitus with an estimated prev- dence of motor dysfunction is less prevalent with
alence ranging up to 50% and possibly higher only 1–6% of diabetic patients displaying clinical
depending on the diagnostic criteria, whether the symptoms and generally occurs in patients with
subjects have type 1 or type 2 diabetes and dura- established diabetic peripheral neuropathy [7]. A
tion of diabetes [1–3]. Diabetic peripheral neu- decrease in motor nerve conduction velocity
ropathy affects both sensorimotor and autonomic early in diabetic animal models is a common
parts of the peripheral nervous system [1]. The finding but there is also evidence in animal mod-
most common clinically recognized form is dia- els and humans of a decrease in compound mus-
betic distal symmetric sensorimotor polyneurop- cle action potential amplitudes and reduced
athy which is characterized by the progressive muscle strength but this has been much less stud-
loss of nerve fibers, both large and small [1, 4]. In ied in the pre-clinical or clinical setting [7, 8].
this progressive disorder the most distal nerve The Diabetes Control and Complications Trial
segments of the feet and hands are affected first (DCCT) and United Kingdom Prospective
and involve retraction of terminal sensory axons Diabetes Study (UKPDS) that focused on type 1
in the periphery with relative preservation of the and type 2 diabetes, respectively, demonstrated
perikarya. This phenomenon is often referred to that hyperglycemia is an important contributing
as “dying back syndrome” or the “stocking and factor to the onset and progression of nerve dam-
glove” pattern reflects damage to the longest sen- age especially in those subjects with type 1 dia-
sory axons first and thus is considered a length-­ betes [9, 10]. However, other reports/studies have
shown that good glycemic control provides little
M. Yorek (*) benefit in towards peripheral neuropathy in those
Department of Internal Medicine, University of Iowa, with type 2 diabetes and recent evidence also
Iowa City, IA, USA suggests that small nerve fiber damage occurs in
Department of Veterans Affairs, Iowa City VA Health individuals with impaired glucose tolerance,
Care System, Iowa City, IA, USA independent of chronic hyperglycemia, and the
Fraternal Order of Eagles Diabetes Research Center, diagnosis of diabetes [11–14]. Thus, other condi-
University of Iowa, Iowa City, IA, USA tions, in addition to hyperglycemia, must contrib-
e-mail: myorek@icva.gov

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 253
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_15
254 M. Yorek

ute to the onset and progression of diabetic 2 Role of Aldose Reductase


peripheral neuropathy in subjects with type 2 dia- and Polyol Pathway
betes. Because of the multiple clinical manifesta-
tions associated with diabetic peripheral The polyol pathway is catalyzed by two enzymes.
neuropathy that can include pain in 15–30% of Aldose reductase, the first enzyme of this path-
diabetic patients with neuropathy determining way, is believed to have a primary role in early
the pathophysiology and therapy for diabetic metabolic damage to peripheral nerves [25]. In
neuropathies is challenging [15]. this initial step glucose is reduced to sorbitol by a
Studies using animal models of both type 1 reaction that requires NADPH, which indirectly
and type 2 diabetes have resulted in the contributes to an increase in oxidative stress. The
identification of wide array of pathological
­ second step is the oxidation of sorbitol to form
mechanisms as contributing to diabetes periph- fructose a reaction catalyzed by NAD-dependent
eral neuropathy. Diabetic peripheral neuropathy sorbitol dehydrogenase (see Fig. 1). During peri-
has been described by some investigators to be a ods of excess glucose such as diabetes tissues
disease of the vasculature leading to nerve isch- independent of insulin for glucose transport
emia and altered nerve function [16–20]. Other accumulate glucose intracellularly leading to an
investigators have proposed that diabetic periph- increased metabolism of glucose by this pathway
eral neuropathy is caused by a combination of and the accumulation of sorbitol causing osmotic
metabolic defects associated with an increased stress and a decrease the intracellular levels of
flux of glucose through the aldose reductase myo-inositol and taurine. The decrease in myo-­
pathway leading to a defect in Na+/K+-ATPase inositol and taurine occurs in response to the
and protein kinase C activities and an alteration excess intracellular accumulation of the osmolyte
of signal transduction pathways in the nerve sorbitol and is referred to the compatible osmo-
[21, 22]. Additional pathologic contributors to lyte hypothesis (to be discussed below). Other
diabetic peripheral neuropathy have been negative consequences of this pathway when
reported to include increased formation of activated are the excess generation of fructose,
advanced glycation endproducts, hexosamine which is a more potent glycation agent than glu-
pathway dysregulation, reduced neurotrophic cose leading to an increase in glycative stress.
support, increased inflammatory, and oxidative The polyol pathway remains an interesting
stress and dyslipidemia [23, 24]. Overall, these therapeutic target for the treatment of diabetic
mechanisms and likely others cause damage to peripheral neuropathy even though early clinical
neurons, Schwann cells, and the vasculature. trials were unsuccessful in achieving meaningful
Ultimately, relentless damage to the nerve com- improvement in human subjects in spite of wide
plex and surrounding vasculature leads to dia- spread success of pre-clinical studies [25–27].
betic peripheral neuropathy. Given the complex Pre-clinical studies with a variety of diabetic ani-
etiology of diabetic peripheral neuropathy a mal models using multiple approaches including
successful treatment will likely require a combi- inhibitors of both aldose reductase and sorbitol
nation of early detection, lifestyle changes, and dehydrogenase and gene and dietary manipula-
pharmaceutical interventions targeting the tion have provided overwhelming evidence of an
mechanisms deemed most responsible for the important role for this pathway in diabetes com-
pathogenesis. Before this can occur additional plications including peripheral neuropathy. In
studies are needed to determine the most rele- rodent studies a wide variety of aldose reductase
vant and targetable causes of diabetic peripheral inhibitors have been shown to improve multiple
neuropathy. endpoints associated with diabetic peripheral
The present review will focus on a number of neuropathy. In this article I will focus on results
the mechanisms introduced above with an from pre-clinical and clinical studies performed
emphasis on those impacted by metabolic with three different aldose reductase inhibitors;
dysregulation. sorbinil, epalrestat and ranirestat.
Metabolic Mechanisms in Diabetic Neuropathy 255

Fig. 1  Illustration of polyol pathway with blue ovals representing conditions that contributes to diabetic peripheral
neuropathy

Sorbinil was one of the earliest aldose reduc- Na+/K+ ATPase activity and myo-inositol con-
tase inhibitors to undergo extensive pre-clinical tent that was corrected following sorbinil treat-
and clinical studies for diabetic peripheral neu- ment [32, 33]. All these studies reported that
ropathy. In studies with type 1 diabetic rats sorbinil treatment reduced tissue sorbitol levels.
treated with streptozotocin treatment with These studies led to the compatible osmolyte
sorbinil prevented as well as reversed defective hypothesis that stated myo-inositol depletion and
axonal transport and slowing of motor nerve con- abnormal signaling by inositol phospholipids
duction velocity [28, 29]. Sorbinil treatment also contribute to a decrease in Na+/K+ ATPase activ-
normalized myo-inositol levels in the sciatic ity in the sciatic nerve, which generates the trans-
nerve in these studies. Another group of investi- membrane sodium and potassium potentials
gators demonstrated that treating streptozotocin-­ necessary for nerve impulse conduction and the
induced diabetic rats with sorbinil corrected both sodium gradient needed for sodium-dependent
motor and sensory nerve conduction velocity as uptake of substrates [32]. The compatible osmo-
well as endoneurial nutritive blood flow of the lyte hypothesis was derived from studies demon-
sciatic nerve, metabolic abnormalities that strating that sorbitol accumulation caused by
included the NAD+/NADH redox imbalance and hyperglycemia/diabetes and activation of the
energy deficiency and improved oxidative stress polyol pathway leads to a corresponding decrease
[30]. Cameron et  al. [31] in studies also using in myo-inositol and taurine levels in peripheral
streptozotocin-diabetic rats demonstrated that nerves [34]. The diabetes-induced decrease in
sorbinil treatment protected axon growth myo-inositol and taurine in peripheral nerves was
retardation. preventable by treatment with an aldose reduc-
In early studies of the sciatic nerve untreated tase inhibitor such as sorbinil that prevented the
diabetic rats were found to have a decrease in increase in sorbitol [35, 36]. This hypothesis as
256 M. Yorek

being a contributing factor to peripheral neuropa- tion are prevented by and aldose reductase inhibi-
thy is supported by additional studies demon- tor [47, 48]. Studies have also been done using an
strating the replenishing myo-inositol or taurine inhibitor of sorbitol dehydrogenase the second
through the diet partially reversed slowing of enzyme in the polyol pathway that converts sor-
motor nerve conduction velocity in diabetic rats bitol to fructose (see Fig.  1). In those studies it
[34, 37, 38]. Using another approach my labora- has been found that blocking sorbitol’s conver-
tory has shown that treating rats with a diet con- sion to fructose exacerbated sympathetic auto-
taining a high concentration of l-fucose caused a nomic neuropathy in streptozotocin-induced
slowing of nerve conduction velocity and diabetic rats and Zucker diabetic rats [49, 50].
decrease sciatic nerve Na+/K+ ATPase activity The effects of the sorbitol dehydrogenase inhibi-
and myo-inositol content [39]. l-Fucose is a tor were prevented by the addition of sorbinil.
potent competitive inhibitor of myo-inositol Lastly, in mice overexpressing human aldose
transport by neural and endothelial cells [40–42]. reductase compared to wild type mice the induc-
Restoring myo-inositol levels through the diet of tion of diabetes using streptozotocin caused a
rats fed the l-fucose diet restored nerve function significantly greater increase in sorbitol and fruc-
and Na+/K+ ATPase activity [39]. My laboratory tose in peripheral nerves even though both sets
has also demonstrated that treating streptozotocin-­ mice had comparable levels of hyperglycemia
induced diabetic rats with sorbinil or myo-­inositol [51]. Both the diabetic wild type and aldose
improved endoneurial blood flow, motor nerve reductase transgenic mice had defective nerve
conduction velocity, and vascular function of epi- conduction velocities but this was significantly
neurial arterioles of the sciatic nerve [43]. The more severe in the diabetic aldose reductase
ladder finding is important because we had previ- transgenic mice. Treating these mice with an
ously demonstrated that decrease of vascular aldose reductase inhibitor significantly prevented
relaxation to acetylcholine by epineurial arteri- the accumulation of sorbitol and slowing of nerve
oles precedes the slowing of nerve conduction conduction velocity [51]. In contrast, studies of
velocity indicating that vascular dysregulation is diabetes in mice deficient in aldose reductase
a contributing factor to diabetes-induced nerve revealed significantly lower levels of sorbitol
dysfunction [44]. Adding to the theory that myo-­ compared to diabetic wild type mice and protec-
inositol depletion contributes to diabetic periph- tion from slowing of nerve conduction velocities
eral neuropathy a recent study has demonstrated [52]. Furthermore, aldose reductase deficiency in
that mRNA and protein expression of myo-­ diabetic mice significantly reduced several mark-
inositol cotransporters in the sciatic nerve are sig- ers of oxidative stress that were significantly
nificantly decreased in experimental diabetes increased in diabetic wild type mice [52].
[45]. The effect of sorbinil has also been exten-
Other animal studies that lend support of the sively studied in human subjects with diabetes.
accumulation of polyols causing slowing of The design of these studies varied as did the num-
motor nerve conduction velocity are galactose ber of subjects enrolled, treatment period, dose,
fed rats. Feeding rats a diet enriched with galac- duration, and endpoints examined. Many of the
tose leads to a large accumulation of the polyol, clinical studies conducted with sorbinil were lim-
galactitol and slowing of motor nerve conduction ited to a small number of subjects and duration of
velocity [46]. Interestingly in this study the treatment of a year or less. The results from these
authors were not able to demonstrate a decrease studies were mixed with some studies reporting a
in sensory nerve conduction velocity with galac- significant improvement in nerve conduction
tose intoxication. In contrast, in streptozotocin-­ velocity and axonal atrophy [53–55] while other
induced diabetic rats both motor and sensory studies reported limited to no benefit [56–58].
nerve conduction velocity was decreased in this The results from these trials was summarized
study [46]. The effects of galactose intoxication nicely by Pfeifer et  al. [59] stating that “future
on polyol accumulation and myo-inositol deple- trials should be designed with adequate statistical
Metabolic Mechanisms in Diabetic Neuropathy 257

power, with consideration of the variability of the effective antioxidant dihydrolipoic acid, which
endpoint measurements for long enough dura- provides a greater protection from oxidative
tion, and with rigorous quality control to defini- damage than does glutathione (GSH) (Fig.  2
tively confirm the utility of aldose reductase [67]). Fidarestat blocked aldose reductase activ-
inhibitors in the treatment of diabetic distal sym- ity thereby conserving NADPH levels, which
metrical polyneuropathy and autonomic neurop- were then available for the production of dihy-
athy.” This review was written in 1997 and the drolipoic acid whose availability is important for
same problems are still a challenge for adequate the production of GSH [67]. Interestingly, several
clinical trials for diabetic peripheral neuropathy groups have reported that the combination of
in 2021. α-lipoic acid and epalrestat is better than mono-
Epalrestat is another aldose reductase inhibi- therapy clinically for diabetic peripheral neurop-
tor that has an extensive history but unlike athy and improvement of motor and sensory
sorbinil it is being used clinically for treatment of nerve conduction velocity [68, 69].
diabetic peripheral neuropathy primarily in Japan Ranirestat is one of the more recent aldose
and recent pre-clinical and clinical studies pro- reductase inhibitors to be tested pre-clinically and
vide evidence that it may also be beneficial for clinically for diabetic peripheral neuropathy.
diabetic nephropathy [60–62]. In pre-clinical Ranirestat is an uncompetitive/reversible inhibitor
studies conducted in rats using a combination of of aldose reductase [70]. In long-term studies
a high fat and high carbohydrate diet followed by with streptozotocin-diabetic rats ranirestat
a low dose of streptozotocin, a model for type 2 reduced sorbitol accumulation in the sciatic nerve
diabetes, it was found that epalrestat treatment and improved the decrease in motor nerve con-
protected the diabetic rats from peripheral neu- duction velocity [71]. Treatment with ranirestat
ropathy through inhibition of the polyol pathway also prevented the deformity of myelinated fibers
and by alleviating oxidative stress [63]. Results and the decrease in their axonal and myelin areas
from several clinical studies report that epalrestat (atrophy) in sural nerves as well as the changes in
treatment of 150 mg/day may improve motor and the size frequency histogram of myelinated fibers
sensory nerve conduction velocity and subjective [70]. In another independent study with a study
neuropathy symptoms with minimal side effects design more relevant to clinical practice ranirestat
[62, 64]. treatment was started 12 weeks after the onset of
In collaboration with the Obrosova laboratory hyperglycemia using streptozotocin-treated rats.
we have also demonstrated that inhibition of At the time of treatment both motor and sensory
aldose reductase with fidarestat of type 1 diabetic nerve conductions were decreased and the
rats attenuates oxidative-nitrosative stress and untreated diabetic rats were hypoalgesic in
activation of poly(ADP-ribose) polymerase while response to a thermal stimulus [72]. Following
improving multiple endpoints associated with only 6 weeks of treatment both motor and sensory
diabetic peripheral neuropathy [65]. This ener- nerve conduction was improved as was the foot
gized studies in my laboratory to examine the withdrawal latency. Ranirestat treatment also
effect of the combination of α-lipoic acid, an improved the intraepidermal nerve fiber density.
antioxidant, and fidarestat on vascular and neural The authors of this study concluded that ranirestat
complications in a type 1 diabetic rat model [66]. has the potential for regeneration in the peripheral
The results from this study demonstrated that the nervous system [72]. Ota et  al. [73] conducted
combination therapy of α-lipoic acid and fidares- studies evaluating the effect of ranirestat com-
tat was more efficacious in preventing diabetes-­ pared to epalrestat treatment on peripheral neu-
induced vascular and neural dysfunction than ropathy and cataract formation in spontaneously
monotherapy that required higher doses to be diabetic torii rats. In this study they found that
equally effective. Our studies attributed this to ranirestat and epalrestat prevented diabetic neu-
the combination therapy allowing for the ropathy but only ranirestat prevented cataract for-
increased conversion of α-lipoic acid to the more mation. There have been several clinical studies
258 M. Yorek

Fig. 2  Lipoic acid (LA) and dihydrolipoic acid (DHLA) metabolism (modified from [67])

performed using ranirestat. Sekiguchi et  al. [74] such as glucose or fructose with free amino
found that 52 weeks of treatment with ranirestat groups of proteins, lipids, or nucleic acids to ini-
(40  mg/day) was well tolerated and improved tially form Schiff bases or Amadori adducts that
nerve conduction velocity but was not able to in the early stages are reversible (Fig. 3) [77–79].
detect any improvement in symptoms and signs. Overtime these products continue to undergo
In two separate studies led by Dr. Vera Bril with reactions that include dehydration, fragmenta-
ranirestat treatment for up to 52–60  weeks with tion, and cross-linking to form irreversible
dosing at 20–40  mg/day it was found treatment advanced glycation endproducts [77]. The accu-
improved motor nerve conduction velocity and mulation of these compounds is associated with
sensory nerve conduction velocity in one study many disease states including diabetes. Their
but not the other [74, 75]. In both studies ranires- accumulation causes structural damage to tissues
tat was reported to be well tolerated with no dif- and organs including components of peripheral
ferences compared to placebo in adverse events. nerves such as Schwann cells and cytoskeletal
Ranirestat is the furthest advanced aldose reduc- proteins; tubulin, neurofilaments, and actin [80,
tase inhibitor for clinical trials except for epalres- 81]. The accumulating damage to these tissues
tat and reproducibly exhibits some degree of culminates in abnormal nerve function including
measurable objective beneficial outcomes [76]. slowed axonal transport, atrophy and degenera-
Its favorable safety profile makes it an attractive tion, and slowing of nerve conduction velocity
choice for further exploration. [77, 82]. Advanced glycation endproducts can
also indirectly affect peripheral nerves by alter-
ing vascular structure and function and ultimately
3 Non-enzymatic Glycation affecting blood flow causing localized ischemia.
Advanced glycation endproducts can also elicit
Another common pathway that was recognized their effects through binding of the receptor for
early to contribute to diabetic peripheral neurop- advanced glycation endproducts (RAGE) [83].
athy was non-enzymatic glycation and the forma- RAGE is expressed in endothelial cells and
tion of advanced glycation endproducts. Schwann cells [84]. Activation of RAGE has
Advanced glycation endproducts are created been shown to stimulate NF-κB and increase oxi-
from non-enzymatic reactions of reducing sugars dative stress [85–87]. Advanced glycation end-
Metabolic Mechanisms in Diabetic Neuropathy 259

Fig. 3  Pathogenic mechanisms associated with non-enzymatic glycation (modified from [77])

products have also been shown to induce diabetic wild type mice with sRAGE with the
basement membrane hypertrophy in endoneurial intent to sequester RAGE ligands also improved
microvessels and disrupt the blood-nerve barrier diabetic peripheral neuropathy [95]. Interestingly,
[88]. In epineurial arterioles, resistance size it has been shown that RAGE is expressed in
blood vessels that provide circulation to the sci- about 30% of all nerve fibers in normal, healthy
atic nerve, endothelium-dependent vascular human subjects, and expression is higher in those
relaxation is mediated in part by acetylcholine subjects with peripheral neuropathy [96].
via nitric oxide generating mechanism important The studies above imply that molecules that
for regulating endoneurial blood flow [89]. We may inhibit the action of advanced glycation end-
and others have demonstrated that endoneurial products may be effective approach for treatment
blood flow is improved with inhibitors of of diabetic peripheral neuropathy. One of the ear-
advanced glycation endproduct formation as well lier compounds used to inhibit the effects of
as oxidative stress; thereby linking these two advanced glycation endproducts was aminogua-
potential mechanisms of diabetes vascular and nidine. Several laboratories including my own
neural complications [43, 90]. Studies with mice have shown that treating diabetic rats with ami-
deficient in RAGE and use of a competitive decoy noguanidine improve peripheral neuropathy with
for advanced glycation endproducts, soluble mechanisms contributing to inhibition of free
RAGE (sRAGE), have contributed to our under- radical formation and improvement in vascular
standing of the role of advanced glycation end- function [22, 43, 97–101]. Pyridoxamine and
products may have in the development of diabetic analogues have also been shown to be effective
peripheral neuropathy [91–93]. In diabetic inhibitors of advanced glycation endproduct for-
RAGE-null mice nerve conduction velocity was mation and treatment for diabetes complications
improved as was nerve generation compared to including diabetic peripheral neuropathy [90,
diabetic wild type mice [94, 95]. Also, treating 102, 103].
260 M. Yorek

Much like the study of the polyol pathway in Protein kinase C (PKC) is another potential tar-
animal models of diabetes there is considerable get for diabetic peripheral neuropathy treatment.
support from pre-clinical studies with diabetic Protein kinase C comprises a superfamily of iso-
rodents for a pathologic role of advanced glyca- enzymes, many of which are activated by 1,2-dia-
tion endproducts in the development and progres- cylglycerol (DAG) [111]. Increased PKC activity
sion of diabetic peripheral neuropathy. is another pathogenic mechanism that has been
Unfortunately, there have been few clinical trials linked to diabetic vascular disease via animal stud-
testing the effects of inhibitors of advanced gly- ies using PKC inhibitors. In the vasculature
cation endproducts on diabetic peripheral increased production of diacylglycerol (DAG)
neuropathy. leads to activation of PKC isoforms (α, β1 and 2,
δ) [111]. Increased PKC activity has been associ-
ated with changes in blood flow, basement mem-
4 Hexosamine and Protein brane thickening, extracellular matrix expansion,
Kinase C increased vascular permeability, and changes in
enzyme activities including Na+-K+ ATPase, phos-
The hexosamine pathway contributes to an phatidylinositol 3-kinase, and mitogen activated
increase in oxidative stress and has been impli- protein kinase [111]. In the nerve DAG levels are
cated in the pathogenesis of diabetes complica- decreased with diabetes and no consistent change
tions including diabetic peripheral neuropathy in activity of PKC isoforms has been observed
[104]. Under conditions of elevated intracellular [111, 112]. Nonetheless, inhibition of PKC β of
glucose and flux into glycolysis, excess, fructose-­ diabetic rats has been reported to correct reduced
6-­phosphate is diverted from glycolysis to pro- nerve blood flow and slowing of nerve conduction
duce glucosamine-6-phosphate, which in turn velocity suggesting a major role of the neurovas-
converts into uridine diphosphate-N-acetyl glu- culature in diabetic peripheral neuropathy [111,
cosamine, a substrate for the formation of proteo- 113–115]. This is further supported by studies
glycans and other glycoproteins [105]. The showing that treatment of diabetic rats with a
diphosphate-N-acetyl glucosamine can also PKCβ inhibitor also improved vascular dysfunc-
increase glycosylation of proteins such as endo- tion of mesenteric arteries to acetylcholine [115].
thelial nitric oxide synthase causing impaired Furthermore, the benefits of PKC inhibition on
enzyme activity, reduced formation of nitric oxide nerve blood flow and nerve conduction velocity
and vascular dysfunction [106]. It is not entirely were attenuated by a nitric oxide synthase inhibi-
clear what kinds of peripheral nerve proteins may tor [116]. Studies of diabetic peripheral neuropa-
be modified by the hexosamine pathway during thy in human subjects treated with a PKC β
diabetes, although there is emerging evidence that inhibitor, ruboxistaurin, for 6  months to 1  year
imbalances in the glycolated state of nervous sys- reported mixed effects with improvement in skin
tem proteins is associated with neurodegenerative microvascular blood flow in diabetes subjects with
disease [107]. One of the more promising treat- neuropathy but limited improvement in sensory
ments based on animal studies of the hexosamine symptoms, vibration detection threshold and
pathway is benfotiamine, a fat-soluble derivative Neuropathy Total Symptom Score-6 [117–119].
of thiamine [108]. In diabetic rats benfotiamine
was found to relieve inflammatory and neuro-
pathic pain [109]. In a phase III trial clinical trial 5 Oxidative and Nitrosative
of 165 patients with symmetrical, distal diabetic Stress
polyneuropathy benfotiamine treatment for
6 weeks reported a significant improvement in the Oxidative stress is a condition resulting from an
Neuropathy Symptom Score with best results imbalance between the generation of reactive
obtained with the high dose according to pain oxygen species (ROS) and the ability of antioxi-
patient-reported symptoms [110]. dant mechanisms to neutralize these compounds
Metabolic Mechanisms in Diabetic Neuropathy 261

[120]. The most common forms of ROS are peripheral neuropathy [124]. Free radicals such
superoxide (O2−), hydrogen peroxide (H2O2), as O2− and OH− cause vascular endothelial dam-
hydroxyl radical (OH−), and peroxynitrite age and reduced NO-mediated vasodilation.
(ONOO−) [121]. Enzymes and pathways located Inhibition of advanced glycosylation and autoxi-
throughout the cell, including the plasma dation, major sources of free radicals, by amino-
membrane, cytosol, mitochondria, and peroxi-
­ guanidine and transition metal chelators, or
somes have the ability to generate these com- antioxidants and free radical scavengers have
pounds under both normal and pathological been demonstrated to improve the diabetes-­
conditions [121]. Superoxide is the most biologi- induced decrease in endoneurial blood flow and
cally important ROS. It can be produced by the improve neural dysfunction such as slowed nerve
electron transport chain of the mitochondria, and conduction velocity [3, 124–132]. The endothe-
by NADH oxidase, NAD(P)H oxidase, xanthine lium, via the release of vasodilators and vasocon-
oxidase, cyclooxygenase, lipoxygenase, cyto- strictors, controls the vascular tone. The three
chrome P-450, and, during periods of tetrahydro- major factors produced by the endothelium that
biopterin deficiency, by nitric oxide synthase contribute to the regulation of vascular relaxation
[121]. Superoxide can spontaneously acquire an are NO, prostacyclin and the as yet an unidenti-
electron to form H2O2. The formation of H2O2 fied factor referred to as endothelium-derived
from O2− can also occur via a reaction catalyzed hyperpolarizing factor (EDHF). Impaired
by superoxide dismutase (SOD) of which there is endothelium-­ dependent vasodilation has been
three isoforms: Mn-SOD, which is located in the demonstrated in various vascular beds of animal
mitochondria and two isoforms of Cu,Zn-SOD, models of diabetes and humans with type 1 and
which are located either in the cytosol or extra- type 2 diabetes [133, 134]. The mechanisms
cellularly [121]. Hydrogen peroxide can be con- induced by hyperglycemia/diabetes considered to
verted to water by the action of catalase or by contribute to endothelial dysfunction as dis-
glutathione peroxidase in the presence of reduced cussed above are the activation of PKC, increased
glutathione [121]. However, in the presence of activity of the polyol pathway, increased forma-
trace metals such as Fe, H2O2 can form OH− via a tion of advanced glycation endproducts, and
process known as the Fenton reaction [121]. The increased oxidative stress. Interestingly, studies
formation of ONOO−, which is the result of a by Brownlee and colleagues have suggested
reaction between O2− and NO, is pathologically hyperglycemia-induced production of O2− by
important in vascular disease and has been dem- mitochondria of endothelial cells as the common
onstrated to be enhanced in diabetes [121, 122]. link for mechanisms of diabetes-induced vascu-
It is ONOO− that is responsible for much of the lar dysfunction [135, 136]. Fernyhough has
cytotoxicity of nitric oxide (NO) that contributes described mitochondrial dysfunction in diabetic
to nitrosative stress [123]. Peroxynitrite has a neuropathy as a “series of unfortunate metabolic
short half-life but is able to diffuse across cell events” [137]. Other chapters in this publication
membranes and depending on the cell environ- will review the impact of mitochondrial dysfunc-
ment can cause a nitrosylation of proteins, which tion, nutrient stress and loss of insulin-dependent
generally reduces enzyme activity, oxidation of growth factor support. Our studies conducted
glutathione, an important antioxidant, and with intact vascular tissue consisting of epineu-
increased peroxidation of lipids [123]. rial arterioles of the sciatic nerve lend support to
As discussed above, endothelial dysfunction the studies by Brownlee and colleagues con-
contributes significantly to diabetic vascular dis- ducted with cultured endothelial cells [138].
ease, which is an important factor in the develop- Studies by my laboratory have provided evidence
ment of diabetic peripheral neuropathy. This is that the generation of oxidative stress through the
supported by studies such as Cameron and Cotter production of O2− and ONOO− impairs vascular
who demonstrated that reduced nerve perfusion function and endothelium-dependent vascular
is a contributing factor in the etiology of diabetic relaxation of epineurial arterioles of the sciatic
262 M. Yorek

nerve from diabetic rats, which precedes the tylcholine [143]. Because metals chelators and
slowing of motor nerve conduction velocity [43, OH− scavengers have also been demonstrated to
44, 125, 139]. Studies designed to investigate the be effective in preventing diabetes-induced vas-
source of O2− formation provided results suggest- cular and neural dysfunction it is likely that the
ing that complex I of the mitochondrial electron formation of OH− may also contribute to impair-
transport chain and possibly NAD(P)H oxidase ment of vascular reactivity and nerve function in
are responsible for the increase in O2− formation diabetes [124, 128, 129, 132, 140, 141, 144].
observed with epineurial arterioles from the sci- As discussed above in regard to improving
atic nerve [138]. It was shown that pretreating diabetes impaired vascular function prevention of
epineurial arterioles from diabetic rats with the oxidative stress is a promising approach for inter-
PKC inhibitor bisindolylmaleimide (GF vention and halting of diabetic peripheral neu-
109203X) improved acetylcholine-mediated vas- ropathy. A variety of antioxidants including
cular relaxation but did not prevent the increase vitamin E have been demonstrated to have bene-
in O2− formation suggesting that activation of ficial effects in treating neuropathy in diabetes
PKC by oxidative stress is downstream of O2− patients and diabetic animal models [145–147].
formation [138]. We have also demonstrated that More recently α-lipoic acid has shown promise
treating diabetic rats with three different types of as a potential antioxidant treatment for diabetic
antioxidants prevented the diabetes-induced neuropathy [148, 149]. Our studies have demon-
increase in O2− and ONOO− formation in aorta strated that α-lipoic acid provides good protec-
and epineurial arterioles of the sciatic nerve and tion against oxidative stress in diabetic rats of
diabetes-induced vascular and neural dysfunc- 4–6  week duration [125]. The treatment of dia-
tion, thereby providing additional evidence that betic rats with α-lipoic acid significantly
increased oxidative stress contributes to diabetes-­ improved diabetes-induced decrease in endoneu-
induced vascular and neural disease [125, 126]. rial blood flow, endothelium-dependent vascular
Studies from other laboratories have provided relaxation in arterioles that provide circulation to
further evidence that antioxidants may prevent the sciatic nerve, and motor nerve conduction
vascular complications in diabetes. Treating dia- velocity. α-Lipoic acid treatment also reduced the
betic rats with tempol, a stable superoxide dis- production of O2− by the aorta and O2− and
mutase mimic compound, abolished the ONOO− by epineurial arterioles. Treating dia-
diabetes-induced increase in vascular O2−, malo- betic rats with α-lipoic acid prevented the
ndialdehyde and 8-epi-prostaglandin F(2α), and diabetes-­induced increase in thiobarbituric acid
also the impairment in relaxation of aortic rings reactive substances in serum and significantly
to acetylcholine [125]. Cameron and colleagues improved lens glutathione levels. α-Lipoic acid is
have demonstrated that treating diabetic rats with a good metal chelator and is capable of scaveng-
α-lipoic acid or the metal chelators hydroxyethyl ing hydroxyl radicals, hypochlorous acid and sin-
starch deferoxamine or trientine prevented the glet oxygen, but not O2− or peroxyl radicals
diabetes-induced impairment in vascular relax- [149–152]. However, in its reduced form, as
ation associated with hyperalgesia and neurovas- dihydrolipoic acid, it is a good scavenger of O2−
cular deficits [128, 129, 140–142]. In addition, and prevents initiation of lipid peroxidation
Keegan et al. demonstrated that treating diabetic [149–152]. In vivo, α-lipoic acid can be con-
rats with α-lipoic acid improved endothelium-­ verted into dihydrolipoic acid [149, 150] (Fig. 2).
dependent vascular relaxation of corpus caverno- This reaction requires NADPH, which is reduced
sum smooth muscle [131]. In another study we in diabetes due to the increased flux of glucose
demonstrated that diabetic peripheral neuropathy through the aldose reductase pathway [120, 153].
in a rat model of type 2 diabetes was improved by Therefore, one potential form of combination
treatment with mitoquinone (Mito-Q) but inter- therapy for the treatment of diabetic neuropathy
estingly in this study treatment did not improve may be combining an aldose reductase inhibitor
vascular reactivity by epineurial arterioles to ace- with α-lipoic acid [66]. This combination should
Metabolic Mechanisms in Diabetic Neuropathy 263

promote the formation of dihydrolipoic acid, smaller molecule (molecular weight 483 vs.
thereby enhancing the antioxidant potential of 30,000 for M40403 and the native enzyme,
α-lipoic acid and possibly providing a synergistic respectively) [126, 158, 159].
effect. In a study by Nakamura et al. of diabetic Another pathway for targeting as a potential
neuropathy in streptozotocin-induced diabetic treatment for diabetic peripheral neuropathy is
rats, they found that treating diabetic rats with the nitrosative stress and the activation of poly(ADP-­
aldose reductase inhibitor NZ-314 improved ribose) polymerase (PARP) [160, 161]. Studies
nerve function and reduced oxidative stress using peroxynitrite decomposition catalysts as
[154]. They concluded that the efficacious effect treatments of diabetic rodents have shown
of aldose reductase inhibition on diabetic neu- improvement in nerve conduction velocity as
ropathy may be mediated by decreasing oxygen well as other sensory related neuropathic deficits
free radicals. This would agree with our studies and improvement of vascular relaxation by epi-
described above demonstrating that treating dia- neurial, coronary, and mesenteric arterioles
betic rats with fidarestat and α-lipoic acid was a [162–165]. These treatments were also shown to
better treatment for diabetic peripheral neuropa- reduce nitrotyrosine and PARP immunofluores-
thy than either monotherapy [66]. Efficacy of cence in the sciatic nerve and dorsal root gan-
α-lipoic acid for diabetic peripheral neuropathy glion neurons [165]. Others studies have
has also been examined in human subjects. In a demonstrated that the PARP inhibitor
review article by Papanas and Ziegler [155] that 3-­aminobenzamide reduced nitrotyrosine immu-
examined placebo-controlled clinical trials and noreactivity in vascular tissue and improved vas-
meta-analyses they concluded that there is evi- cular reactivity of epineurial arterioles and neural
dence supporting α-lipoic acid as an efficacious function [166–168]. Studies from my laboratory
and safe treatment for diabetic neuropathy. that lend support to PARP activation and meta-
The development of superoxide dismutase bolic stress contributing to diabetic peripheral
mimetics are another class of antioxidants with neuropathy have shown that treating dietary
potential for treatment of diabetes complications obese or type 2 diabetic mice with nicotinamide
including diabetic peripheral neuropathy [126, riboside improved multiple endpoints associated
156, 157]. Because of limitations associated with with peripheral neuropathy [169].
enzyme therapies these non-peptidyl compounds
may offer advantages resulting in better clinical
therapies and outcomes for diseases mediated by 6 Additional Mechanisms
O2− radicals such as diabetes [158]. In our studies and Treatment Options
we demonstrated that treating diabetic rats with
M40403 inhibited the generation of O2− by aorta Other pathways and downstream mediators of
and epineurial vessels of the sciatic nerve, the pathology associated with diabetic peripheral
formation of ONOO− by epineurial vessels of the neuropathy include mitogen activated protein
sciatic nerve, the reduction in endoneurial blood kinase (MAPK), nuclear factor κB (NF-κB),
flow, the slowing of motor nerve conduction cyclooxygenase 2 (COX-2), 12/15-lipoxygenase
velocity and impairment of endothelium-­ (12/15-LOX), and Na+/H+-exchanger-1 (NHE-1).
dependent vasodilation of epineurial arterioles. It These downstream mediators of neural impair-
also improved the diabetes induced increase in ment have in common their activation via oxida-
serum TBARS and sciatic nerve conjugated diene tive/inflammatory stress. Activation of the MAPK
level, two additional markers of oxidative stress pathway has been documented in neuron and
[126]. M40403 is a prototypic example of a sta- Schwann cells by hyperglycemia as well as in
ble, low molecular weight, manganese-­sciatic nerve of diabetic rodents and human sub-
containing, non-peptidic molecule possessing the jects with type 1 and type 2 diabetes [170, 171].
function and catalytic rate of native SOD Different treatment modalities have been
enzymes, but with the advantage of being a much employed in pre-clinical studies for treatment of
264 M. Yorek

diabetic peripheral neuropathy showing improve- the sciatic nerve and spinal cord of diabetic
ment in neural endpoints as well as reduction in rodents [190]. Pharmacological inhibition of
activation of MAPK [172–174]. These studies 12/15-lipoxygenase or gene deficiency has been
indicate a potential therapeutic approach for shown to alleviate some but not all pathological
treatment of diabetic peripheral neuropathy changes associated with diabetic peripheral neu-
through inhibition of MAPK pathway. ropathy of large and small fibers while reducing
Activation of PARP and MAPK can contrib- MAPK activation and relieving oxidative and
ute to regulation of gene expression through acti- inflammatory stress [191–193].
vation of NF-κB [175]. Nuclear factor-2 erythroid In recent years my laboratory has focused on
related factor-2 (Nrf2) and NF-κB pathways are two additional pathways of intervention as a ther-
potential therapeutic targets for diabetic neuropa- apeutic target for diabetic peripheral neuropathy.
thy [176]. Pre-clinical studies therapeutically tar- In the next two sections I will briefly review our
geting Nrf2 and NF-κB have demonstrated by studies of the effect of inhibition of neutral endo-
inhibiting NF-κB and/or activating Nrf2 can peptidase or neprilysin and nutritional supple-
reduce inflammation and oxidative stress and mentation of omega-3 polyunsaturated fatty acids
improve endpoints related to diabetic peripheral (PUFA) primarily found in fish oil eicosapentae-
neuropathy [177–182]. noic acid (EPA) and docosahexaenoic acid
Other potential targets for treatment of dia- (DHA) and their metabolites. I have previously
betic peripheral neuropathy include inhibition of provided reviews for both of these topics [194,
Na+/K+ exchangers [183]. Treating streptozotocin-­ 195].
induced diabetic rats with cariporide reversed Angiotensin converting enzyme (ACE),
multiple endpoints associated with diabetic dipeptidyl peptidase IV (DPP-IV), and neprilysin
peripheral neuropathy and interestingly also (neutral endopeptidase) are all proteases. As a
reduced markers linked to increased oxidative group these proteases can contribute to disease
and nitrosative stress and advanced glycation conditions through the breakdown of important
endproducts suggesting that the pathology of vasoactive and neuroprotective peptides as well
activation of Na+/K+ exchangers in diabetes as other peptides such as glucagon-like peptide 1.
occurs upstream of these common mediators of DPP-IV inhibitors (gliptins) are a class of drugs
diabetic peripheral neuropathy. approved by the FDA for hyperglycemia-related
Impairment in lipid metabolism is another treatment of type 2 diabetes [196]. Pre-clinical
potential therapeutic target for treatment of dia- studies have demonstrated that dipeptidyl pepti-
betic peripheral neuropathy [184]. dase IV inhibitors are also an effective treatment
Cyclooxygenase-2 pathway is upregulated in the for diabetic peripheral neuropathy [197–200].
peripheral nerves and dorsal root ganglia neurons The renin-angiotensin system (RAS) plays a
in experimental diabetes and chemical inhibition major role in the pathophysiology of cardiovas-
or gene deletion improves peripheral nerve func- cular and renal disease [201–203]. Most known
tion including nerve conduction velocity and loss effects of angiotensin II are mediated via activa-
of epidermal nerve fibers while reducing oxida- tion of the AT1-receptor [202]. Activation of the
tive stress and inflammation [185, 186]. Treating AT1-receptor is involved in vasoconstriction,
diabetic mice with the cyclooxygenase-2 inhibi- inactivation of bradykinin, water and salt homeo-
tor meloxicam has been shown to reduce neuro- stasis, control of neuro-humoral systems, reac-
pathic pain [187]. Other anti-inflammatory or tive oxygen species production, cellular
anti-oxidative stress reagents thymoquinone and hypertrophy, and hyperplasia and apoptosis
sulforaphane have been shown to improve in pre-­ [203]. ACE inhibitors and angiotensin receptor
clinical studies diabetic peripheral neuropathy blockers are well-known treatments for hyperten-
while reducing cyclooxygenase-2 expression in sion, atherosclerosis, congestive heart failure,
Schwann cells or sciatic nerves [188, 189]. stroke, and myocardial infarction including those
12/15-Lipoxygenase overexpression is evident in patients with diabetes [201, 204]. In diabetic ani-
Metabolic Mechanisms in Diabetic Neuropathy 265

mal models we and others have demonstrated diabetic peripheral neuropathy until we per-
that ACE inhibitors and to a lesser extent angio- formed our studies. Using the vasopeptidase
tensin receptor blockers can alleviate many defi- inhibitor ilepatril (AVE7688) we demonstrated in
cits associated with diabetic peripheral both pre-diabetic and type 1 or type 2 diabetic
neuropathy as well as vascular dysfunction of mice and rats that the combined inhibition of
epineurial arterioles of the sciatic nerve [205– ACE and neprilysin was more effective than
210]. In addition to pre-clinical studies in animal monotherapy in improving neurovascular func-
models of diabetes there are also results from tion and slowing/reversing motor and sensory
three small clinical trials demonstrating that dia- peripheral nerve deficits including diabetes-­
betic neuropathy can be improved by treatment induced decrease in innervation and sensitivity of
of patients with quinapril, trandolapril or lisino- corneal nerves [207, 230–234]. Furthermore, we
pril [211–213]. demonstrated that deletion of the neprilysin gene
The other lesser known protease that is protected mice from the development of neuro-
included in this discussion is neprilysin. pathologic deficits and loss of corneal nerves
Neprilysin is found in many tissues including upon induction of diabetes [235]. However,
vascular and renal tissue and its activity is despite greater efficacy for hypertension than
increased by fatty acids and glucose in human monotherapy, development of vasopeptidase
microvascular cells [214–217]. Interestingly, inhibitors was halted due to significant off-target
neprilysin activity has been shown to be activated effects in some cohorts, most notably increased
by protein kinase C, which is increased in vascu- frequency of angioedema in hypertensive sub-
lar tissues by diabetes including endothelial cells jects [236]. This led to the development of
[218, 219]. Importantly for this discussion nepri- LCZ696 by Novartis a combination of sacubitril
lysin degrades vasoactive neuropeptides includ- (neprilysin inhibitor) and valsartan (angiotensin
ing; natriuretic peptides, adrenomedullin, receptor blocker) (Entresto). In 2015 the FDA
bradykinin, endothelin, glucagon-like peptide 1, approved the use of sacubitril/valsartan in
substance P, and calcitonin gene-related peptide patients with heart failure with reduced ejection
[220, 221]. Thus, inhibition of this protease pro- fraction [237]. We obtained this drug to study its
vides great potential for the treatment of diabetes effect on vascular and neural complications in a
complications. To test this question several phar- rat model of type 2 diabetes [238]. Using an early
maceutical companies created what was termed and late intervention study design the results
vasopeptidase inhibitors. At the time these inhibi- demonstrated efficacy of sacubitril/valsartan in
tors were a new class of drug designed to simul- improving vascular and neural function that was
taneously inhibit neprilysin and ACE activity superior to valsartan alone. In the early interven-
[222]. They were first developed for treating tion protocol, sacubitril/valsartan treatment was
hypertension and heart failure [223, 224]. In found to slow progression of these deficits and,
regard to diabetes complications it has been with late intervention treatment, was found to
shown that vascular conductance in the femoral stimulate restoration of vascular reactivity, motor
artery of streptozotocin-induced diabetic rats was and sensory nerve conduction velocities, and sen-
improved by a vasopeptidase inhibitor [225]. sitivity/regeneration of sensory nerves of the skin
Furthermore, it has been demonstrated that vaso- and cornea. We concluded that sacubitril/valsar-
peptidase inhibitors are neuroprotective and pre- tan may be an effective treatment for diabetic
vent nephropathy in ZDF rats [226, 227]. peripheral neuropathy and due to its safety profile
Vasopeptidase inhibitors have also been reported and FDA approval could advance quickly to clin-
to decrease matrix metalloproteinases and AGE ical trials.
accumulation/formation in type 2 diabetes and Recently we have also been interested in the
improve wound healing [228, 229]. However, potential for omega-3 PUFA as a treatment for
there was no information available about the diabetic peripheral neuropathy. Since inflamma-
potential benefits of vasopeptidase inhibitors in tory stress is considered to be a primary mecha-
266 M. Yorek

nism for diabetic peripheral neuropathy and were effective in reversing impaired nerve conduc-
omega-3 PUFA and their metabolites are known tion velocities and nerve fiber density and sensitiv-
to have anti-inflammatory properties it was rea- ity in the skin and cornea [255–257]. Furthermore,
sonable to expect that fish oil, a common source our studies have demonstrated that E and D series
for omega-3 PUFA, may be an effective treat- resolvins (resolution-phase interaction products),
ment [239, 240]. Although trials using treatment metabolites of EPA and DHA, respectively, (see
with omega-3 PUFA have primarily focused on Fig.  4) which are known to have anti-inflamma-
cardiovascular disease risk reduction [241–246], tory and neuroprotective properties, may be the
there is significant evidence of their benefits active mediator of the EPA and DHA effects on
beyond cardiovascular disease. For instance, sev- diabetic peripheral neuropathy and nerve regener-
eral recent studies have reported potential bene- ation [255, 258, 259]. Thus, optimizing production
fits of omega-3 PUFA consumption on several of these metabolites may be an effective approach
chronic inflammatory and autoimmune diseases, to increasing the efficacy of EPA and DHA sup-
stroke, muscle atrophy, and neurodegenerative plements in the diet, and thus ensuring a better out-
disease [247–254]. Nonetheless, the consump- come for diabetic peripheral neuropathy. In that
tion of omega-3 PUFA remains low in the regard we have demonstrated that the combination
Western diet due to historically increased con- of menhaden oil and salsalate, a highly effective
sumption of omega-6 PUFA. agent in blocking proinflammatory chemokines
In our own most recent pre-clinical studies, we and cytokines, with a large margin of safety and
found that long-chain omega-3 PUFA, primarily low costs [260, 261] increase production of resol-
EPA and DHA derived from fish (menhaden) oil, vin D1 in  vivo vs. menhaden oil alone and this

Elcosapentaenoic acid Docosahexaenoic acid


(EPA, 20:5n-3) (DHA, 22:6n-3)
COOH
COOH

3 3
COX-2/Aspirin CX-2/Aspirin 12-LOX
CYP450 15-lipoxgenase

Maresins
18-HEPE
Maresins 1 OH

5-LOX 15-LOX
COOH
Resolvin
E series
OH
Resolvin E1 Resolvin E3
Protectins
OH OH Protectins D1
COOH
COOH
5-LOX COOH

OH OH OH HO OH
Resolvin D series
Resolvin E2
Resolvin D1
OH
HO OH
COOH
COOH

OH
OH
Metabolism of EPA and DHA

Fig. 4  Metabolism of EPA and DHA


Metabolic Mechanisms in Diabetic Neuropathy 267

combination is more efficacious toward improving has expanded considerably. However, this infor-
peripheral neuropathy in diabetic rodents [259, mation has not provided us with a successful
262]. Another important evidence, supporting treatment. There are numerous reasons why this
future clinical trials of omega-3 PUFA is that our may be the case with some of this being covered
pre-­clinical studies have found that the circulating in other chapters. From my point of view some
levels of EPA and DHA in rats receiving menha- of the reasons that may factor into these past fail-
den oil was comparable to the EPA and DHA ures are that animals are not suitable models for
serum levels of human subjects taking 4  g of diabetic neuropathy as it occurs in humans,
EPA + DHA per day (fish oil capsules) for 4 weeks many of the human studies that have been con-
[257, 263]. For instance, the omega-3 PUFA levels ducted to date are flawed with patients being to
in serum increased from 5.6% to 14.4% and the advance in disease, treatment periods to short
EPA to AA ratio increased from 0.12 to 0.88  in and endpoints not adequate to detect early status
human participants treated with 4 g of fish oil for of disease and reversibility. However, a primary
4 weeks [263]. This is similar to the 3.9% to 15.4% reason I believe for the failure of finding an
increase in the serum omega-3 PUFA levels and effective treatment for diabetic peripheral neu-
0.04 to 0.54 increase in the EPA to AA ratio we ropathy is the monotherapy approach that has
found in our study of diabetic rats treated with been taken for many of the studies. It has been
menhaden oil [256]. Thus, increasing the circula- repeatedly demonstrated and reported that the
tory levels of EPA and DHA, and favorably alter- etiology of diabetic peripheral neuropathy is
ing the omega-3 index to a healthy range of 8–12% complex with many mechanisms, some dis-
could be an effective disease modifying therapy cussed in this chapter as well as in other chap-
for treating diabetic peripheral neuropathy [264]. ters, contributing to the development and
In humans, a proof-of-concept study that progression of the disease. Our cardiology col-
tested the effects of 12  months of ~2  g daily leagues routinely treat their patients with hyper-
omega-3 PUFA supplementation on the progres- tension by advising them to consider lifestyle
sion of diabetic peripheral neuropathy in patients and dietary changes as well as prescribing sev-
with type 1 diabetes, reported a 29% significant eral different drugs that target the different
increase in corneal nerve fiber length (primary potential mechanisms responsible for high blood
outcome), but no effect on sensory nerve conduc- pressure. Thus, it seems irrational to think that
tion velocity [265]. Unfortunately, it is unclear if anything other than a combination of lifestyle
omega-3 PUFA therapeutic levels were achieved changes that include increased exercise and bet-
in these participants given that serum omega-3 ter dietary choices and a combination of inter-
index or omega-3 PUFA metabolites were not ventional drugs will be needed to successfully
assessed. However, the above change in corneal treat diabetic peripheral neuropathy. I have pro-
fiber length was comparable to the increase in vided you evidence of this from different animal
corneal fiber length we observed in type 1 diabe- studies in this chapter. What investigators and
tes rats treated with menhaden oil and provides clinicians need to be asking are what the primary
encouraging information to support future clini- targets are for successfully treating diabetic
cal trials using omega-3 PUFA and salsalate as an peripheral neuropathy and what the best drugs to
intervention for diabetic peripheral neuropathy use for these targets are?
[256].

References
7 Conclusion
1. Zenker J, Ziegler D, Chrast R.  Novel pathogenic
pathways in diabetic neuropathy. Trends Neurosci.
Over the years our knowledge and understanding 2013;36:439–49.
of the different mechanisms that contribute to 2. Vinik A.  Clinical practice. Diabetic sensory and
the etiology of diabetic peripheral neuropathy motor neuropathy. N Engl J Med. 2016;374:1455–64.
268 M. Yorek

3. Pop-Busui R, Boulton A, Feldman E, et  al. and streptozotocin-­ diabetic rats. Diabetologia.
Diabetic neuropathy: a position statement by the 1994;37:449–59.
American Diabetes Association. Diabetes Care. 20. Kles KA, Vinik AI. Pathophysiology and treatment
2017;40:136–54. of diabetic peripheral neuropathy: the case for dia-
4. Perkins B, Bril V.  Electrophysiology test- betic neurovascular function as an essential compo-
ing in diabetic neuropathy. Handb Clin Neurol. nent. Curr Diabetes Rev. 2006;2:131–45.
2014;126:235–48. 21. Cotter MA, Dines KC, Cameron NE.  Prevention
5. Feldman E, Callaghan B, Pop-Busui R, et al. Diabetic and reversal of motor and sensory peripheral nerve
neuropathy. Nat Rev Dis Primers. 2019;5:41. conduction abnormalities in streptozotocin-diabetic
6. Kobayashi M, Zochodne DW.  Diabetic neuropathy rats by the prostacyclin analogue iloprost. Naunyn
and the sensory neuron: new aspects of pathogenesis Schmiedebergs Arch Pharm. 1993;347:534–40.
and their treatment implications. J Diabetes Investig. 22. Cameron NE, Cotter MA, Dines KC, Love A. Effects
2018;9:1239–54. of aminoguanidine on peripheral nerve function
7. Wilson NM, Wright DE.  Experimental motor and polyol pathway metabolites in streptozotocin-­
neuropathy in diabetes. Handb Clin Neurol. diabetic rats. Diabetologia. 1992;35:946–50.
2014;126:461–7. 23. Sima AA.  Pathological mechanisms involved in
8. Zochodne DW, Ramji N, Toth C. Neuronal targeting diabetic neuropathy: can we slow the process? Curr
in diabetes mellitus: a story of sensory neurons and Opin Investig Drugs. 2006;7:324–37.
motor neurons. Neuroscientist. 2008;14:311–8. 24. Pop-Busui R, Sima A, Stevens M. Diabetic neuropa-
9. Writing team for the Diabetes Control and thy and oxidative stress. Diabetes Metab Res Rev.
Complications Trial/Epidemiology of Diabetes 2006;22:257–73.
Interventions and Complications Research Group. 25. Gabbay KH. Aldose reductase inhibition in the treat-
Effect of intensive therapy on the microvascular ment of diabetic neuropathy: where are we in 2004?
complications of type 1 diabetes mellitus. JAMA. Curr Diab Rep. 2004;4:405–8.
2002;287:2563–9. 26. Oates PJ.  Aldose reductase, still a compelling tar-
10. Stratton I, Adler AI, Neil H, et  al. Association get for diabetic neuropathy. Curr Drug Targets.
of glycaemia with macrovascular and microvas- 2008;9:14–36.
cular complications of type 2 diabetes (UKPDS 27. Chalk C, Benstead TJ, Moore F.  Aldose reductase
35): prospective observational study. BMJ. inhibitors for the treatment of diabetic polyneuropa-
2000;321:405–12. thy. Cochrane Database Syst Rev. 2007;CD004572.
11. Bönhof GJ, Herder C, Strom A, Papanas N, Roden 28. Tomlinson DR, Moriarty RJ, Mayer JH. Prevention
M, Ziegler D. Emerging biomarkers, tools, and treat- and reversal of defective axonal transport and motor
ments for diabetic polyneuropathy. Endocr Rev. nerve conduction velocity in rats with experimen-
2019;40:153–92. tal diabetes by treatment with the aldose reductase
12. Zochodne D.  Diabetes mellitus and the peripheral inhibitor Sorbinil. Diabetes. 1984;33:470–6.
nervous system: manifestations and mechanisms. 29. Willars GB, Tomlinson DR, Robinson JP.  Studies
Muscle Nerve. 2007;36:144–66. of sorbinil on axonal transport in streptozotocin-­
13. Tavakoli M, Mojaddidi M, Fadavi H, Malik diabetic rats. Metabolism. 1986;35(4 Suppl
R. Pathophysiology and treatment of painful diabetic 1):66–70.
neuropathy. Curr Pain Headache Rep. 2008;12:192–7. 30. Obrosova IG, Huysen CV, Fathallah L, Cao XC,
14. Smith A, Singleton J.  Impaired glucose tolerance Greene DA, Stevens MJ.  As aldose reductase
and neuropathy. Neurologist. 2008;14:23–9. inhibitor reverses early diabetes-induced changes in
15. Malik R. Wherefore art thou, O treatment for diabetic peripheral nerve function, metabolism, and antioxi-
neuropathy? Int Rev Neurobiol. 2016;127:287–317. dative defense. FASEB J. 2002;16:123–5.
16. Cameron NE, Cotter MA, Low PA.  Nerve blood 31. Cameron NE, Leonard MB, Ross IS, Whiting
flow in early experimental diabetes in rats: relation PH. The effects of sorbinil on peripheral nerve con-
to conduction deficits. Am J Phys. 1991;261:E1–8. duction velocity, polyol concentrations and morphol-
17. Nukada H, Dyck PJ.  Microsphere embolization ogy in the streptozotocin-diabetic rat. Diabetologia.
of nerve capillaries and fiber degeneration. Am J 1986;29:168–74.
Pathol. 1984;115:275–87. 32. Greene DA, Lattimer SA.  Recent advances in
18. Cameron NE, Cotter MA, Dines KC, Maxfield the therapy of diabetic peripheral neuropathy by
EK, Carey F, Mirrlees DJ. Aldose reductase inhibi- means of a aldose reductase inhibitor. Am J Med.
tion, nerve perfusion, oxygenation and function in 1985;79:13–7.
streptozotocin-diabetic rats: dose-response consider- 33. Greene DA.  A sodium-pump defect in diabetic
ations and independence from a myo-inositol mech- peripheral nerve corrected by sorbinil administra-
anism. Diabetologia. 1994;37:651–63. tion: relationship to myo-inositol metabolism and
19. Cameron NE, Cotter MA, Archibald V, Dines nerve conduction slowing. Metabolism. 1986;35(4
KC, Maxfield EK.  Anti-oxidant and pro-oxidant Suppl 1):60–5.
effects on nerve conduction velocity, endoneu- 34. Stevens MJ, Lattimer SA, Kamijo M, Van Huysen
rial blood flow and oxygen tension in non-diabetic C, Sima AA, Greene DA.  Osmotically-induced
Metabolic Mechanisms in Diabetic Neuropathy 269

nerve taurine depletion and the compatible osmolyte 47. Dvornik E, Simard-Duquesne N, Krami M, et  al.
hypothesis in experimental diabetic neuropathy in Polyol accumulation in galactosemic and dia-
the rat. Diabetologia. 1993;36:608–14. betic rats: control by an aldose reductase inhibitor.
35. Greene DA, Mackway AM. Decreased myo-­inositol Science. 1973;182:1146–8.
content and Na+-K+-ATPase activity in superior 48. Kamijo M, Basso M, Cherian PV, Hohman TC, Sima
cervical ganglion of STZ-diabetic rat and pre- AA. Galactosemia produces ARI-preventable nodal
vention by aldose reductase inhibition. Diabetes. changes similar to those of diabetic neuropathy.
1986;35:1106–8. Diabetes Res Clin Pract. 1194;25:117–29.
36. Finegold D, Lattimer SA, Nolle S, Bernstein M, 49. Schmidt RE, Dorsey DA, Beaudet LN, et  al. A
Greene DA.  Polyol pathway activity and myo-­ potent sorbitol dehydrogenase inhibitor exacer-
inositol metabolism. A suggested relationship in bates sympathetic autonomic neuropathy in rats
the pathogenesis of diabetic neuropathy. Diabetes. with streptozotocin-induced diabetes. Exp Neurol.
1983;32:988–92. 2005;192:407–19.
37. Gillon KR, Hawthorne JN, Tomlinson DR. Myo-­ 50. Schmidt RE, Dorsey DA, Beaudet LN, et  al.
inositol and sorbitol metabolism in relation to Inhibition of sorbitol dehydrogenase exacerbates
peripheral nerve function in experimental diabe- autonomic neuropathy in rats with streptozotocin-­
tes in the rat: effect of aldose reductase inhibition. induced diabetes. J Neuropathol Exp Neurol.
Diabetologia. 1983;25:365–71. 2001;60:1153–69.
38. Pop-Busui R, Sullivan KA, Van Huysen CV, et  al. 51. Yagihashi S, Yamagishi SI, Wada R, et al. Neuropathy
Depletion of taurine in experimental diabetic neurop- in diabetic mice overexpressing human aldose reduc-
athy: implications for nerve metabolic, vascular, and tase and effects of aldose reductase inhibitor. Brain.
functional deficits. Exp Neurol. 2001;168:259–72. 2001;124:2448–58.
39. Yorek MA, Wiese TJ, Davidson EP, et al. Reduced 52. Ho ECM, Lam KSL, Chen YS, et  al. Aldose
motor nerve conduction velocity and Na(+)-K(+)- reductase-­deficient mice are protected from delayed
ATPase activity in rats maintained on L-fucose diet. motor nerve conduction velocity, increased c-Jun
Reversal by myo-inositol supplementation. Diabetes. NH2-terminal kinase activation, depletion of reduced
1993;42:1401–6. glutathione, increased superoxide accumulation, and
40. Yorek MA, Stefani MR, Dunlap JA, Ro KS, DNA damage. Diabetes. 2006;55:1946–53.
Davidson EP.  Trans-hydroxyl group configuration 53. Sima AA, Bril V, Nathaniel V, et  al. Regeneration
on carbons 2 and 3 of glucose. Responsible for and repair of myelinated fibers in sural-nerve
acute inhibition of myo-inositol transport? Diabetes. biopsy specimens from patients with diabetic
1991;40:1016–23. neuropathy treated with sorbinil. N Engl J Med.
41. Yorek MA, Dunlap JA, Stefani MR, Davidson 1988;319:548–55.
EP.  L-fucose is a potent inhibitor of myo-inositol 54. Sima AA, Prashar A, Nathaniel V, Bril V, Werb MR,
transport and metabolism in cultured neuroblastoma Greene DA.  Overt diabetic neuropathy: repair of
cells. J Neurochem. 1992;58:1626–36. axo-glial dysjunction and axonal atrophy by aldose
42. Stefani MR, Dunlap JA, Yorek MA.  Effect of reductase inhibition and its correlation to improve-
L-fucose on proliferation and myo-inositol metabo- ment in nerve conduction velocity. Diabet Med.
lism in cultured cerebral microvessel and aortic 1993;10:115–21.
endothelial cells. J Cell Physiol. 1992;153:321–31. 55. Gieron MA, Malone JI, Lowitt S, Korthals
43. Coppey LJ, Gellett JS, Davidson EP, Dunlap JA, JK. Improvement in peripheral nerve function after
Yorek MA. Effect of treating streptozotocin-induced one year of Sorbinil. Neuroreport. 1991;2:348–50.
diabetic rats with sorbinil, myo-inositol or amino- 56. Lewin IG, O’Brien IA, Morgan MH, Corrall
guanidine on endoneurial blood flow, motor nerve RJ. Clinical and neurophysiological studies with the
conduction velocity and vascular function of epineu- aldose reductase inhibitor, sorbinil, in symptomatic
rial arterioles of the sciatic nerve. Int J Diabetes Res. diabetic neuropathy. Diabetologia. 1984;26:445–8.
2002;3:21–36. 57. O’Hare JP, Morgan MH, Alden P, Chissel S, O’Brien
44. Coppey LJ, Davidson EP, Dunlap JA, Lund DD, IA, Corrall RJ.  Aldose reductase inhibition in dia-
Yorek MA.  Slowing of motor nerve conduction betic neuropathy: clinical and neurophysiological
velocity in streptozotocin-induced diabetic rats is studies of one year’s treatment with sorbinil. Diabet
preceded by impaired vasodilation in arterioles that Med. 1988;5:537–42.
overlie the sciatic nerve. Int J Exp Diabetes Res. 58. Fagius J, Brattberg A, Jameson S, Berne C. Limited
2000;1:131–43. benefit of treatment of diabetic polyneuropathy with
45. Farias VX, Uchoa PN, Aquino CP, et al. Expression an aldose reductase inhibitor: a 24-week controlled
of myo-inositol cotransporters in the sciatic nerve trial. Diabetologia. 1985;28:323–9.
and dorsal root ganglia in experimental diabetes. 59. Pfeifer MA, Schumer MP, Gelber DA.  Aldose
Braz J Med Biol Res. 2019;52:e8589. reductase inhibitors: the end of an era or the need
46. Kalichman MW, Dines KC, Bobik M, Mizisin for different trial designs? Diabetes. 1997;46(Suppl
AP.  Nerve conduction velocity, laser Doppler flow, 2):S82–9.
and axonal caliber in galactose and streptozotocin 60. He J, Gao H-X, Yang N, et al. The aldose reductase
diabetes. Brain Res. 1998;810:130–7. inhibitor epalrestat exerts nephritic protection on dia-
270 M. Yorek

betic nephropathy in db/db mice through metabolic ception, and intraepidermal nerve fiber density in
modulation. Acta Pharmacol Sin. 2019;40:86–97. rats with overt diabetic polyneuropathy. J Diabetes
61. Hotta N, Kawamori R, Fukuda M, Shigeta Y. Long-­ Res. 2019;2019:2756020.
term clinical effects of epalrestat, an aldose reduc- 73. Ota A, Kakehashi A, Toyoda F, et al. Effects of long-­
tase inhibitor, on progression of diabetic neuropathy term treatment with ranirestat, a potent aldose reduc-
and other microvascular complications: multivariate tase inhibitor, on diabetic cataract and neuropathy in
epidemiological analysis based on patient back- spontaneously diabetic torii rats. J Diabetes Res.
ground factors and severity of diabetic neuropathy. 2013;2013:175901.
Diabet Med. 2012;29:1529–33. 74. Sekiguchi K, Kohara N, Baba M, et al. Aldose reduc-
62. Ramirez MA, Borja NL. Epalrestat: an aldose reduc- tase inhibitor ranirestat significantly improves nerve
tase inhibitor for the treatment of diabetic neuropa- conduction velocity in diabetic polyneuropathy: a
thy. Pharmacotherapy. 2008;28:646–55. randomized double-blind placebo-controlled study
63. Li Q-R, Wang Z, Zhou W, et al. Epalrestat protects in Japan. J Diabetes Investig. 2019;10:466–74.
against diabetic peripheral neuropathy by alleviat- 75. Bril V, Hirose T, Tomioka S, Buchanan R. Ranirestat
ing oxidative stress and inhibiting polyol pathway. for the management of diabetic sensorimotor poly-
Neural Regen Res. 2016;11:345–51. neuropathy. Diabetes Care. 2009;32:1256–60.
64. Hotta N, Akanuma Y, Kawamori R, et al. Long-term 76. Giannoukakis N.  Evaluation of ranirestat for the
clinical effects of epalrestat, an aldose reductase treatment of diabetic neuropathy. Expert Opin Drug
inhibitor, on diabetic peripheral neuropathy: the Metab Toxicol. 2014;10:1051–9.
3-year, multicenter, comparative Aldose Reductase 77. Sima AAF, Sugimoto K.  Experimental dia-
Inhibitor-Diabetes Complications Trial. Diabetic betic neuropathy: an update. Diabetologia.
Care. 2006;29:1538–44. 1999;42:773–88.
65. Obrosova IG, Pacher P, Szabo C, et al. Aldose reduc- 78. Brownlee M, Cerami A, Vlassara H. Advanced gly-
tase inhibition counteracts oxidative-nitrosative cosylation end products in tissue and the biochemi-
stress and poly(ADP-ribose) polymerase activation cal basis of diabetic complications. N Engl J Med.
in tissue sites for diabetes complications. Diabetes. 1988;318:1315–21.
2005;54:234–42. 79. Zochodne D. Mechanisms of diabetic neuron dam-
66. Yorek MA, Coppey LJ, Gellett JS, Davidson EP, age: molecular pathways. Handb Clin Neurol.
Lund DD. Effect of fidarestat and alpha-lipoic acid 2014;126:379–99.
on diabetes-induced epineurial arteriole vascular 80. Ryle C, Leow CK, Donaghy M.  Nonenzymatic
dysfunction. Exp Diabesity Res. 2004;5:123–35. glycation of peripheral and central nervous system
67. Rochette L, Ghibu S, Muresan A, Vergely C. Alpha-­ proteins in experimental diabetes mellitus. Muscle
lipoic acid: molecular mechanisms and therapeu- Nerve. 1997;20:577–84.
tic potential in diabetes. Can J Physiol Pharmacol. 81. Cullum NA, Mahon J, Stringer K, McLean
2015;93:1021–7. WG.  Glycation of rat sciatic nerve tubulin in
68. Wang X-T, Lin H-X, Xu S-A, Lu Y-K. Lipoic acid experimental diabetic mellitus. Diabetologia.
combined with epalrestat versus lipoic acid in 1991;34:387–9.
treating diabetic peripheral neuropathy: a meta-­ 82. Ozturk G, Sekeroglu MR, Erdogan E, Ozturk M. The
analysis. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. effect of non-enzymatic glycation of extracellular
2017;39:656–64. matrix proteins on axonal regeneration in vitro. Acta
69. Zhao M, Chen J-Y, Chu Y-D, Zhu Y-B, Luo L, Bu Neuropathol. 2006;112:627–32.
S-Z. Efficacy of epalrestat plus α-lipoic acid combi- 83. Forbes J, Cooper M. Mechanisms of diabetic com-
nation therapy versus monotherapy in patients with plications. Physiol Rev. 2013;93:137–88.
diabetic peripheral neuropathy: a meta-analysis of 84. Wada R, Yagihashi S.  Role of advanced glyca-
20 randomized controlled trials. Neural Regen Res. tion end products and their receptors in develop-
2018;13:1087–95. ment of diabetic neuropathy. Ann N Y Acad Sci.
70. Matsumoto T, Ono Y, Kurono M, Kuromiya A, 2005;1043:598–604.
Nakamura K, Bril V. Ranirestat (AS-3201), a potent 85. Bekircan-Kurt CB, Tan E, Ozdamar SE.  The acti-
aldose reductase inhibitor, reduces sorbitol lev- vation of RAGE and NF-κB in nerve biopsies of
els and improves motor nerve conduction velocity patients with axonal and vasculitic neuropathy. Noro
in streptozotocin-diabetic rats. J Pharmacol Sci. Psikiyatr Ars. 2015;52:279–82.
2008;107:231–7. 86. Yamagishi S, Nakamura K, Matsui T, Noda Y,
71. Matsumoto T, Ono Y, Kurono M, Kuromiya A, Imaizumi T.  Receptor for advanced glycation end
Toyosawa K, Ueda Y, Bril V.  Long-term treat- products (RAGE): a novel therapeutic target for
ment with ranirestat (AS-3201), a potent aldose diabetic vascular complication. Curr Pharm Des.
reductase inhibitor, suppresses diabetic neuropathy 2008;14:487–95.
and cataract formation in rats. J Pharmacol Sci. 87. Yamagishi S, Nakamura K, Matsui T, Ueda S,
2008;107:340–8. Fukami K, Okuda S.  Agents that block advanced
72. Asano S, Himeno T, Hayami T, et  al. Ranirestat glycation end product (AGE)-RAGE (receptor for
improved nerve conduction velocities, sensory per- AGEs)-oxidative stress system: a novel therapeutic
Metabolic Mechanisms in Diabetic Neuropathy 271

strategy for diabetic vascular complications. Expert 102. Metz TO, Alderson NL, Thorpe SR, Baynes
Opin Investig Drugs. 2008;17:983–96. JW. Pyridoxamine, an inhibitor of advanced glyca-
88. Shimizu F, Sano Y, Haruki H, Kanda T.  Advanced tion and lipoxidation reactions: a novel therapy for
glycation end-products induce basement mem- treatment of diabetic complications. Arch Biochem
brane hypertrophy in endoneurial microvessels Biophys. 2003;419:41–9.
and disrupt the blood-nerve barrier by stimulat- 103. Nagai R, Schirakawa JI, Ohno RI, Moroishi N,
ing the release of TGF-β and vascular endothelial Nagai M.  Inhibition of AGEs formation by natural
growth factor (VEGF) by pericytes. Diabetologia. products. Amino Acids. 2014;46:261–6.
2011;54:1517–26. 104. Du XL, Edelstein D, Rossetti L, et al. Hyperglycemia-­
89. Terata K, Coppey LJ, Davidson EP, Dunlap JA, induced mitochondrial superoxide overproduction
Gutterman DD, Yorek MA.  Acetylcholine-induced activates the hexosamine pathway and induces plas-
arteriolar dilation is reduced in streptozotocin-­ minogen activator inhibitor-1 expression by increas-
induced diabetic rats with motor nerve dysfunction. ing Sp1 glycosylation. Proc Natl Acad Sci U S A.
Br J Pharmacol. 1999;128:837–43. 2000;97:12222–6.
90. Cameron NE, Gibson TM, Nangle MR, Cotter 105. Román-Pintos LM, Villegas-Rivera G, Rodríguez-­
MA.  Inhibitors of advanced glycation end prod- Carrizalez AD, Miranda-Díaz AG, Cardona-­
uct formation and neurovascular dysfunction Muñoz EG.  Mellitus: inflammation, oxidative
in experimental diabetes. Ann N Y Acad Sci. stress, and mitochondrial function. J Diabetes Res.
2005;1043:784–92. 2016;3425617
91. Sugimoto K, Yasujima M, Yagihashi S.  Role of 106. Yagihashi S.  Glucotoxic mechanisms and related
advanced glycation end products in diabetic neu- therapeutic approaches. Int Rev Neurobiol.
ropathy. Curr Pharm Des. 2008;14:953–61. 2016;127:121–49.
92. Lukic I, Humpert P, Nawroth P, Bierhaus A.  The 107. Heath JM, Sun Y, Yuan K, et al. Activation of AKT
RAGE pathway: activation and perpetuation in the by O-linked N-acetylglucosamine induces vas-
pathogenesis of diabetic neuropathy. Ann N Y Acad cular calcification in diabetes mellitus. Circ Res.
Sci. 2008;1126:76–80. 2014;114:1094–102.
93. Toth C, Martinez J, Zochodne D.  RAGE, dia- 108. Hammes H, Du X, Edelstein D, et al. Benfotiamine
betes, and the nervous system. Curr Mol Med. blocks three major pathways of hyperglycemic dam-
2007;7:766–76. age and prevents experimental diabetic retinopathy.
94. Juranek JK, Geddis MS, Song F, et al. RAGE defi- Nat Med. 2003;9:294–9.
ciency improves postinjury sciatic nerve regeneration 109. Sanchez-Ramirez GM, Caram-Salas NL, Rocha-­
in type 1 diabetic mice. Diabetes. 2013;62:931–43. Ganzalez HI, et al. Benfotiamine relieves inflamma-
95. Bierhaus A, Humpert PM, Morcos M, et  al. tory and neuropathic pain in rats. Eur J Pharmacol.
Understanding RAGE, the receptor for advanced 2006;530:48–53.
glycation end products. J Mol Med. 2005;83:876–86. 110. Stracke H, Gaus W, Achenbach U, Federlin K,
96. Juranek JK, Kothary P, Mehra A, Hays A, Brannagan Bretzel R.  Benfotiamine in diabetic polyneuropa-
TH, Schmidt AM. Increased expression of the recep- thy (BEN-DIP): results of a randomized, double-­
tor for advanced glycation end-products in human blind, placebo-controlled clinical study. Exp Clin
peripheral neuropathies. Brain Behav. 2013;3:701–9. Endocrinol Diabetes. 2008;116:600–5.
97. Yagihashi S, Kamijo M, Baba M, Yagihashi N, Nagai 111. Eichberg J.  Protein kinase C changes in diabe-
K.  Effect of aminoguanidine on functional and tes: is the concept relevant to neuropathy? Int Rev
structural abnormalities in peripheral nerve of STZ-­ Neurobiol. 2002;50:61–82.
induced diabetic rats. Diabetes. 1992;41:47–52. 112. Evcimen ND, King GL. The role of protein kinase C
98. Miyauchi Y, Shikama H, Takasu T, et al. Slowing of activation and the vascular complications of diabe-
peripheral motor nerve conduction was ameliorated tes. Pharmacol Res. 2007;55:498–510.
by aminoguanidine in streptozotocin-induced dia- 113. Sasase T, Morinaga H, Abe T, et al. Protein kinase C
betic rats. Eur J Endocrinol. 1996;134:467–73. beta inhibitor prevents diabetic peripheral neuropa-
99. Sugimoto K, Yagihashi S. Effects of aminoguanidine thy, but not histopathological abnormalities of retina
on structural alterations of microvessels in periph- in spontaneously diabetic Torii rat. Diabetes Obes
eral nerve of streptozotocin diabetic rats. Microvasc Metab. 2009;11:1084–7.
Res. 1997;53:105–12. 114. Nakamura J, Kato K, Hamada Y, et  al. A protein
100. Kihara M, Schmelzer JD, Poduslo JF, Curran GL, kinase C-beta-selective inhibitor ameliorates neural
Nickander KK, Low PA.  Aminoguanidine effects dysfunction in streptozotocin-induced diabetic rats.
on nerve blood flow, vascular permeability, elec- Diabetes. 1999;48:2090–5.
trophysiology, and oxygen free radicals. Proc Natl 115. Cotter MA, Jack AM, Cameron NE.  Effects of the
Acad Sci U S A. 1991;88:6107–11. protein kinase C beta inhibitor LY333531 on neural
101. Cameron NE, Cotter MA. Rapid reversal by amino- and vascular function in rats with streptozotocin-­
guanidine of the neurovascular effects of diabetes in induced diabetes. Clin Sci. 2002;103:311–21.
rats: modulation by nitric oxide synthase inhibition. 116. Cameron NE, Cotter MA, Jack AM, Basso MD,
Metabolism. 1996;45:1147–52. Hohman TC. Protein kinase C effects on nerve func-
272 M. Yorek

tion, perfusion, Na(+),K(+)-ATPase activity and transition metal chelating agents. J Clin Invest.
glutathione content in diabetic rats. Diabetologia. 1995;96:1159–63.
1999;42:1120–30. 130. Cameron NE, Cotter MA.  Effects of antioxidants
117. Casellini CM, Barlow PM, Rice AL, et  al. A on nerve and vascular dysfunction in experimental
6-month randomized, double-blind-masked, diabetes. Diabetes Res Clin Pract. 1999;45:137–46.
placebo-­controlled study evaluating the effects of 131. Keegan A, Cotter MA, Cameron NE. Effects of dia-
the protein kinase C-beta inhibitor ruboxistaurin on betes and treatment with the antioxidant α-lipoic acid
skin microvascular blood flow and other measures on endothelial and neurogenic responses of corpus
of diabetic peripheral neuropathy. Diabetes Care. cavernosum in rats. Diabetologia. 1999;42:343–50.
2007;30:896–902. 132. Karasu C, Dewhurst M, Stevens EJ, Tomlinson
118. Brooks B, Delaney-Robinson C, Molyneaux L, DR.  Effects of anti-oxidant treatment on sciatic
Yue DK.  Endothelial and neural regulation of skin nerve dysfunction in streptozotocin-diabetic rats;
microvascular blood flow in patients with diabetic comparison with essential fatty acids. Diabetologia.
peripheral neuropathy: effect of treatment with the 1995;38:129–34.
isoform-specific protein kinase C beta inhibitor, 133. Pieper GM, Siebeneich W. Diabetes-induced endo-
ruboxistaurin. J Diabetes Complicat. 2008;22:88–95. thelial dysfunction is prevented by long-term treat-
119. Vinik AI, Bril V, Kempler P, et  al. Treatment of ment with the modified iron chelator, hydroxyethyl
symptomatic diabetic peripheral neuropathy with starch conjugated-deferoxamine. J Cardiovasc
the protein kinase C beta-inhibitor ruboxistaurin Pharmacol. 1997;30:734–8.
mesylate during a 1-year randomized, placebo-­ 134. De Vriese AS, Verbeuren TJ, Van de Voorde J,
controlled, double-blind clinical trial. Clin Ther. Lameire NH. Vanhoutte PM endothelial dysfunction
2005;27:1164–80. in diabetes. Br J Pharmacol. 2000;130:963–74.
120. van Dam PS.  Oxidative stress and diabetic neu- 135. Brownlee M.  Biochemistry and molecular cell
ropathy: pathophysiological mechanisms and biology of diabetic complications. Nature.
treatment perspectives. Diabetes Metab Res Rev. 2001;414:813–20.
2002;18:176–84. 136. Nishikawa T, Edelstein D, Du XL, et al. Normalizing
121. Schnackenberg CG.  Physiological and pathophysi- mitochondrial superoxide production blocks three
ological roles of oxygen radicals in the renal micro- pathways of hyperglycaemic damage. Nature.
vasculature. Am J Phys. 2002;282:R335–42. 2000;404:787–90.
122. Radi R, Peluffo G, Alvarez MN, Naviliat M, Cayota 137. Fernyhough P.  Mitochondrial dysfunction in dia-
A. Unraveling peroxynitrite formation in biological betic neuropathy: a series of unfortunate metabolic
systems. Free Radic Biol Med. 2001;30:463–88. events. Curr Diab Rep. 2015;15:89.
123. Fredstrom S.  Nitric oxide, oxidative stress, and 138. Coppey LJ, Gellett JS, Davidson EP, Yorek
dietary antioxidants. Nutrition. 2002;18:537–9. MA.  Preventing superoxide formation in epineu-
124. Cameron NE, Cotter MA.  Metabolic and vascular rial arterioles of the sciatic nerve from diabetic rats
factors in the pathogenesis of diabetic neuropathy. restores endothelium-dependent vasodilation. Free
Diabetes. 1997;46:S31–7. Radic Res. 2003;37:33–40.
125. Coppey LJ, Gellett JS, Davidson EP, Dunlap JA, 139. Nassar T, Kadery B, Lotan C, Da’as N, Kleinman Y,
Lund DD, Yorek MA.  Effect of antioxidant treat- Haj-Yehia A.  Effects of the superoxide dismutase-­
ment of streptozotocin-induced diabetic rats on mimetic compound tempol on endothelial dysfunc-
endoneurial blood flow, motor nerve conduc- tion in streptozotocin-induced diabetic rats. Eur J
tion velocity and vascular reactivity of epineu- Pharmacol. 2002;436:111–8.
rial arterioles of the sciatic nerve. Diabetes. 140. Cameron NE, Jack AM, Cotter MA.  Effect of
2001;50:1927–37. alpha-­lipoic acid on vascular responses and noci-
126. Coppey LJ, Gellett JS, Davidson EP, et  al. Effect ception in diabetic rats. Free Radic Biol Med.
of M40403 treatment of diabetic rats on endoneu- 2001;31:125–35.
rial blood flow, motor nerve conduction velocity and 141. Cameron NE, Cotter MA. Effects of an extracellular
vascular function of epineurial arterioles of the sci- metal chelator on neurovascular function in diabetic
atic nerve. Br J Pharmacol. 2001;134:21–9. rats. Diabetologia. 2001;44:621–8.
127. Yorek MA, Coppey LJ, Gellett JS, et al. Effect of treat- 142. Inkster ME, Cotter MA, Cameron NE. Effects of tri-
ment of diabetic rats with Dehydroepiandrosterone entine, a metal chelator, on defective endothelium-­
(DHEA) on vascular and neural function. Am J dependent relaxation in the mesenteric vasculature
Phys. 2002;283:E1067–75. of diabetic rats. Free Radic Res. 2002;36:1091–9.
128. Cameron NE, Cotter MA, Maxfield EK. Antioxidant 143. Fink B, Coppey L, Davidson E, et al. Effect of mito-
treatment prevents the development of peripheral quinone (Mito-Q) on neuropathic endpoints in an
nerve dysfunction in streptozotocin-diabetic rats. obese and type 2 diabetic rat model. Free Radic Res.
Diabetologia. 1993;36:299–304. 2020;54:311–8.
129. Cameron NE, Cotter MA.  Neurovascular dys- 144. Cameron NE, Tuck Z, McCabe L, Cotter MA. Effect
function in diabetic rats: potential contribution of the hydroxyl radical scavenger, dimethylthiourea,
of autoxidation and free radicals examined using on peripheral nerve tissue perfusion, conduction
Metabolic Mechanisms in Diabetic Neuropathy 273

velocity and nociception in experimental diabetes. genesis of diabetic complications. Emerging


Diabetologia. 2001;44:1161–9. new therapeutical strategies. Curr Med Chem.
145. Manzella D, Barbieri M, Ragno E, Paolisso 2005;12:267–75.
G.  Chronic administration of pharmacologic doses 161. Cowell RM, Russell JW. Nitrsative injury and anti-
of vitamin E improves the cardiac autonomic ner- oxidant therapy in the management of diabetic neu-
vous system in patients with type 2 diabetes. Am J ropathy. J Investig Med. 2004;52:33–44.
Clin Nutr. 2001;73:1052–7. 162. Vareniuk I, Pavlov IA, Drel VR, et  al. Nitrosative
146. van Dam PS, Bravenboer B, van Asbeck BS, Marx stress and peripheral diabetic neuropathy in
JJ, Gispen WH.  High rat food vitamin E content leptin-deficient (ob/ob) mice. Exp Neurol.
improves nerve function in streptozotocin-diabetic 2007;205:425–36.
rats. Eur J Pharmcol. 1999;376:217–22. 163. Obrosova IG, Drel VR, Oltman CL, et  al. Role of
147. Nicklander KK, Schmelzer JD, Rohwer DA, Low nitrosative stress in early neuropathy and vascular
PA. Effect of alpha-tocopherol deficiency on indices dysfunction in streptozotocin-diabetic rats. Am J
of oxidative stress in normal and diabetic peripheral Physiol Endocrinol Metab. 2007;293:E1645–55.
nerve. J Neurol Sci. 1994;126:6–14. 164. Obrosova IG, Mabley JG, Zsengeller Z, et al. Role
148. Kocak G, Aktan F, Canbolat O, et  al. Alpha-lipoic for nitrosative stress in diabetic neuropathy: evi-
acid treatment ameliorates metabolic parameters, dence from studies with a peroxynitrite decomposi-
blood pressure, vascular reactivity and morphol- tion catalyst. FASEB J. 2005;19:401–3.
ogy of vessels already damaged by streptozotocin-­ 165. Drel VR, Pacher P, Vareniuk I, et  al. A peroxyni-
diabetes. Diabetes Nutr Metab. 2000;13:308–18. trite decomposition catalyst counteracts sensory
149. Coleman MD, Eason RC, Bailey CJ. The therapeutic neuropathy in streptozotocin-diabetic mice. Eur J
use of lipoic acid in diabetes: a current perspective. Pharmacol. 2007;569:48–58.
Environ Toxicol Pharmacol. 2001;10:167–72. 166. Obrosova IG, Drel VR, Pacher P, et  al. Oxidative-­
150. Packer L, Kraemer K, Rimbach G.  Molecular nitrosative stress and poly(ADP-ribose) poly-
aspects of lipoic acid in the prevention of diabetes merase (PARP) activation in experimental diabetic
complications. Nutrition. 2001;17:888–95. neuropathy: the relation is revisited. Diabetes.
151. Dincer Y, Telci A, Kayah R, Yilmaz IA, Cakatay U, 2005;54:3435–41.
Akcay T. Effect of α-lipoic acid on lipid peroxida- 167. Obrosova IG, Li F, Abatan OI, et  al. Role of
tion and anti-oxidant enzyme activities in diabetic poly(ADP-ribose) polymerase activation in diabetic
rats. Clin Exp Pharmacol Physiol. 2002;29:281–4. neuropathy. Diabetes. 2004;53:711–20.
152. Jones W, Li X, Zhi-Chao Q, Perriott L, Whitesell 168. Ilnytska O, Lyzogubov VV, Stevens MJ, et  al.
RR, May JM.  Uptake, recycling , and antioxidant Poly(ADP-ribose) polymerase inhibition alleviates
actions of α-lipoic acid in endothelial cells. Free Rad experimental diabetic sensory neuropathy. Diabetes.
Biol Med. 2002;33:83–93. 2006;55:1686–94.
153. Bravenboer B, Kappelle AC, Hamers FPR, van 169. Trammell SAJ, Weidemann BJ, Chadda A, et  al.
Buren T, Erkelens DW, Gispen WH.  Potential use Nicotinamide riboside opposes type 2 diabetes and
of glutathione for the prevention and treatment of neuropathy in mice. Sci Rep. 2016;6:26933.
diabetic neuropathy in the streptozotocin-induced 170. Purves T, Middlemas A, Agthong S, et al. A role for
diabetic rat. Diabetologia. 1992;35:813–7. mitogen-activated protein kinases in the etiology of
154. Nakamura J, Hamada Y, Chaya S, et  al. Transition diabetic neuropathy. FASEB J. 2001;15:2508–14.
metals and polyol pathway in the development of 171. Zhu H, Yu W-J, Le Y, et  al. High glucose levels
diabetic neuropathy in rats. Diabetes Metab Res Rev. increase the expression of neurotrophic factors
2002;18:395–402. associated with p-p42/p44 MAPK in Schwann cells
155. Papanas N, Ziegler D.  Efficacy of α-lipoic acid in in vitro. Mol Med Rep. 2012;6:179–84.
diabetic neuropathy. Expert Opin Pharmacother. 172. Stavniichuk R, Drel VR, Shevalye H, et  al.
2014;15:2721–31. Baicalein alleviates diabetic peripheral neuropa-
156. Cuzzocrea S, Riley DP, Caputi AP, Salvemini thy through inhibition of oxidative-nitrosative
D.  Antioxidant therapy: a new pharmacological stress and p38 MAPK activation. Exp Neurol.
approach in shock, inflammation, and ischemia/ 2011;230:106–13.
reperfusion injury. Pharmacol Rev. 2001;53:135–59. 173. Zhou J, Du X, Long M, et al. Neuroprotective effect
157. Doggrell SA.  Therapeutic potential of selective of berberine is mediated by MAPK signaling path-
superoxide dismutase mimetics. Drugs Future. way in experimental diabetic neuropathy in rats. Eur
2002;27:385–90. J Pharmacol. 2016;774:87–94.
158. Salvemini D, Wang ZQ, Zweier JL, et al. A nonpep- 174. Sweitzer SM, Medicherla S, Almirez R, et  al.
tidyl mimic of superoxide dismutase with therapeu- Antinociceptive action of a p38alpha MAPK inhibi-
tic activity in rats. Science. 1999;286:304–6. tor, SD-282, in a diabetic neuropathy model. Pain.
159. Salvemini D, Riley DP.  M40403. Drugs Fut. 2004;109:409–19.
2002;25:1027–33. 175. Kovacs K, Vaczy A, Fekete K, et  al. PARP inhibi-
160. Pacher P, Obrosova IG, Mabley JG, Szabo C. Role tor protects against chronic hypoxia/reoxygenation-­
of nitrosative stress and peroxynitrite in the patho- induced retinal injury by regulation of MAPKs,
274 M. Yorek

HIF1α, Nrf2, and NFκB. Invest Ophthalmol Vis Sci. 189. Moustafa P, Abdelkader NF, El-Awdan SA,
2019;60:1478–90. El-Shabrawy OA, Zaki HF.  Extracellular matrix
176. Yerra VG, Negi G, Sharma SS, Kumar A. Potential remodeling and modulation of inflammation and
therapeutic effects of the simultaneous targeting of oxidative stress by sulforaphane in experimen-
the Nrf2 and NF-κB pathways in diabetic neuropa- tal diabetic peripheral neuropathy. Inflammation.
thy. Redox Biol. 2013;1:394–7. 2018;41:1460–76.
177. Li J, Hu X, Liang F, et  al. Therapeutic effects of 190. Stavniichuk R, Drel VR, Shevalye H, et al. Role of
moxibustion simultaneously targeting Nrf2 and 12/15-lipoxygenase in nitrosative stress and periph-
NF-κB in diabetic peripheral neuropathy. Appl eral prediabetic and diabetic neuropathies. Free
Biochem Biotechnol. 2019;189:1167–82. Radic Biol Med. 2010;49:1036–45.
178. Shay KP, Moreau RF, Smith EJ, Smith AR, Hagen 191. Stavniichuk R, Obrosov AA, Drel VR, Nadler JL,
TM.  Alpha-lipoic acid as a dietary supplement: Obrosova IG, Yorek MA. 12/15-Lipoxygenase inhi-
molecular mechanisms and therapeutic potential. bition counteracts MAPK phosphorylation in mouse
Biochim Biophys Acta. 2009;1790:1149–60. and cell culture models of diabetic peripheral neu-
179. Agca CA, Tuzcu M, Hayirli A, Sahin K.  Taurine ropathy. J Diabetes Mellitus. 2013;3:33015. https://
ameliorates neuropathy via regulating NF-κB and doi.org/10.4236/jdm.2013.33015.
Nrf2/HO-1 signaling cascades in diabetic rats. Food 192. Obrosova IG, Stavniichuk R, Drel VR, et  al.
Chem Toxicol. 2014;71:116–21. Different roles of 12/15-lipoxygenase in diabetic
180. Ma J, Shi M, Zhang X, et al. GLP-1R agonists ame- large and small fiber peripheral and autonomic neu-
liorate peripheral nerve dysfunction and inflamma- ropathies. Am J Pathol. 2010;177:1436–47.
tion via p38 MAPK/NF-κB signaling pathways in 193. Stavniichuk R, Shevalye H, Hirooka H, Nadler JL,
streptozotocin-induced diabetic rats. Int J Mol Med. Obrosova IG. Interplay of sorbitol pathway or glu-
2018;41:2977–85. cose metabolism, 12/15-lipoxygenase, and mitogen-­
181. Kumar A, Negi G, Sharma SS.  Suppression of activated protein kinase in the pathogenesis of
NF-κB and NI-κB regulated oxidative stress and diabetic peripheral neuropathy. Biochem Pharmacol.
neuroinflammation by BAY11-7082 (IκB phosphor- 2012;83:932–40.
ylation inhibitor) in experimental diabetic neuropa- 194. Yorek MA.  The potential role of angiotensin con-
thy. Biochimie. 2012;94:1158–65. verting enzyme and vasopeptidase inhibitors in the
182. Kumar A, Sharma SS.  NF-kappaB inhibitory treatment of diabetic neuropathy. Curr Drug Targets.
action of resveratrol: a probable mechanism 2008;9:77–84.
of neuroprotection in experimental diabetic 195. Yorek MA. Is fish oil a potential treatment for dia-
neuropathy. Biochem Biophys Res Commun. betic peripheral neuropathy? Curr Diabetes Rev.
2010;394:360–5. 2018;14:339–49.
183. Lupachyk S, Watcho P, Shevalye H, et  al. Na+/ 196. Deacon CF.  Dipeptidyl peptidase 4 inhibitors in
K+ exchanger 1 inhibition reverses manifesta- the treatment of type 2 diabetes mellitus. Nat Rev
tion of peripheral diabetic neuropathy in type 1 Endocrinol. 2020;16:642–53.
diabetic rats. Am J Physiol Endocrinol Metab. 197. Jin HY, Liu WJ, Park JH, Baek HS, Park
2013;305:E396–404. TS.  Effect of dipeptidyl peptidase-IV (DPP-IV)
184. Kellogg AP, Cheng HT, Pop-Busui inhibitor (Vildagliptin) on peripheral nerves in
R. Cyclooxygenase-2 pathway as a potential thera- streptozotocin-­induced diabetic rats. Arch Med Res.
peutic target in diabetic peripheral neuropathy. Curr 2009;40:536–44.
Drug Targets. 2008;9:68–76. 198. Tsubi K, Mizukami H, Inaba W, Baba M, Yagihashi
185. Kellogg AP, Pop-Busui R. Peripheral nerve dysfunc- S. The dipeptidyl peptidase IV inhibitor vildagliptin
tion in experimental diabetes is mediated by cyclo- suppresses development of neuropathy in diabetic
oxygenase-­2 and oxidative stress. Antioxid Redox rodents: effects on peripheral sensory nerve func-
Signal. 2005;7:1521–9. tion, structure and molecular changes. J Neurochem.
186. Kellogg AP, Wiggin TD, Larkin DD, Hayes JM, 2016;136:859–70.
Stevens MJ, Pop-Busui R.  Protective effects of 199. Bianchi R, Cervellini C, Porretta-Serapiglia C, et al.
cyclooxygenase-2 gene inactivation against periph- Beneficial effects of PKF275-055, a novel, selec-
eral nerve dysfunction and intraepidermal nerve tive, orally bioavailable, long-lasting dipeptidyl
fiber loss in experimental diabetes. Diabetes. peptidase IV inhibitor in streptozotocin-induced dia-
2007;56:2997–3005. betic peripheral neuropathy. J Pharmacol Exp Ther.
187. Kimura S, Kontani H.  Demonstration of antial- 2012;340:64–72.
lodynic effects of the cyclooxygenase-2 inhibitor 200. Davidson EP, Coppey LJ, Dake B, Yorek
meloxicam on established diabetic neuropathic pain MA.  Treatment of streptozotocin-induced dia-
in mice. J Pharmacol Sci. 2009;110:213–7. betic rats with alogliptin: effect on vascular
188. Chen L, Li B, Chen B, et  al. Thymoquinone alle- and neural complications. Exp Diabetes Res.
viates the experimental diabetic peripheral neu- 2011;2011:810469.
ropathy by modulation of inflammation. Sci Rep. 201. Volpe M, Savoia C, De Paolis P, Ostrowska B, Tarasi
2016;6:31656. D, Rubattu S. The renin-angiotensin system as a risk
Metabolic Mechanisms in Diabetic Neuropathy 275

factor and therapeutic target for cardiovascular and peptidase activity in human microvascular endothe-
renal disease. J Am Soc Nephrol. 2002;13:S173–8. lial cells. Shock. 2003;19:508–12.
202. Nickenig G, Ostergren J, Struijker-Boudier 216. Edwards RM, Pullen M, Nambi P.  Distribution of
H. Clinical evidence for the cardiovascular benefits of neutral endopeptidase activity along the rat and rab-
angiotensin receptor blockers. J Renin-Angiotensin-­ bit nephron. Pharmacology. 1999;59:45–50.
Aldosterone Syst. 2006;7(Suppl 1):S1–7. 217. Gonzalez W, Soleilhac JM, Fournie-Zaluski MC,
203. Mogensen CE. Microalbuminuria and hypertension Roques BP, Michel JB.  Characterization of neu-
with focus on type 1 and type 2 diabetes. J Intern tral endopeptidase in vascular cells, modulation
Med. 2003;254:45–66. of vasoactive peptide levels. Eur J Pharmacol.
204. Li EC, Heran BS, Wright JM. Angiotensin converting 1998;345:323–31.
enzyme (ACE) inhibitors versus angiotensin recep- 218. Kikkawa F, Shibata K, Suzuki T, et  al. Signal
tor blockers for primary hypertension. Cochrane pathway involved in increased expression of neu-
Database Syst Rev. 2014;2014:CD009096. tral endopeptidase 24.11 by gonadotropin releas-
205. Cameron NE, Cotter MA, Robertson S. Angiotensin ing hormone in choriocarcinoma cells. Placenta.
converting enzyme inhibition prevents develop- 2004;25:176–83.
ment of muscle and nerve dysfunction and stimu- 219. Suzki T, Ino K, Kikkawa F, et al. Neutral endopepti-
lates angiogenesis in streptozotocin-diabetic rats. dase/CD10 expression during phorbol ester-induced
Diabetologia. 1992;35:12–8. differentiation of choriocarcinoma cells through the
206. Coppey LJ, Davidson EP, Rinehart TW, et al. ACE protein kinase C- and extracellular signal-regulated
inhibitor or angiotensin II receptor antagonist atten- kinase-dependent signalling pathway. Placenta.
uates diabetic neuropathy in streptozotocin-induced 2002;23:475–82.
diabetic rats. Diabetes. 2006;55:341–8. 220. Pu Q, Schiffrin EL.  Effect of ACE/NEP inhibition
207. Davidson EP, Coppey LJ, Holmes A, Yorek on cardiac and vascular collagen in stroke-prone
MA.  Effect of inhibition of angiotensin convert- spontaneously hypertensive rats. Am J Hypertens.
ing enzyme and/or neutral endopeptidase on vas- 2001;14:1067–72.
cular and neural complications in high fat fed/low 221. Plamboeck A, Holst JJ, Carr RD, Deacon CF. Neutral
dose streptozotocin-diabetic rats. Eur J Pharmacol. endopeptidase 24.11 and dipeptidyl peptidase IV are
2012;677:180–7. both mediators of the degradation of glucagon-like
208. Manschot SM, Gispen WH, Kappelle LJ, Biessels peptide 1  in the anaesthetised pig. Diabetologia.
GJ.  Nerve conduction velocity and evoked poten- 2005;48:1882–90.
tial latencies in streptozotocin-diabetic rats: effects 222. Weber M.  Emerging treatments for hypertension:
of treatment with an angiotensin converting enzyme potential role for vasopeptidase inhibition. Am J
inhibitor. Diabetes Metab Res Rev. 2003;19:469–77. Hypertens. 1999;12:139S–47S.
209. Aggarwal M, Singh J, Sood S, Arora B.  Effects of 223. Trindade PT, Rouleau JL. Vasopeptidase inhibitors:
lisinopril on streptozotocin-induced diabetic neu- potential role in the treatment of heart failure. Heart
ropathy in rats. Methods Find Exp Clin Pharmacol. Fail Monit. 2001;2:2–7.
2001;23:131–4. 224. Nathisuwan S, Talbert RL. A review of vasopeptidase
210. Oltman CL, Davidson EP, Coppey LJ, Kleinschmidt inhibitors: a new modality in the treatment of hyper-
TL, Lund DD, Yorek MA. Attenuation of vascular/ tension and chronic heart failure. Pharmacotherapy.
neural dysfunction in Zucker rats treated with enala- 2002;22:27–42.
pril or rosuvastatin. Obesity. 2008;16:82–9. 225. Arbin V, Claperon N, Fournie-Zaluski MC, Roques
211. Didangelos T, Tziomalos K, Margaritidis C, et  al. BP, Peyroux J.  Effects of combined neutral endo-
Efficacy of administration of an angiotensin convert- peptidase 24-11 and angiotensin-converting enzyme
ing enzyme inhibitor for two years on autonomic and inhibition on femoral vascular conductance in
peripheral neuropathy in patients with diabetes mel- streptozotocin-­induced diabetic rats. Br J Pharmacol.
litus. J Diabetes Res. 2017;2017:6719239. 2000;130:1297–304.
212. Malik RA, Williamson S, Abbott C, et  al. Effect 226. Schafer S, Schmidts H-L, Bleich M, Busch AE,
of angiotensin-converting-enzyme (ACE) inhibi- Linz W.  Nephroprotection in Zucker diabetic fatty
tor trandolapril on human diabetic neuropathy: rats by vasopeptidase inhibition is partly brady-
randomized double-blind controlled trial. Lancet. kinin B2 receptor dependent. Br J Pharmacol.
1998;352:1978–81. 2004;143:27–32.
213. Reja A, Tesfaye S, Harris ND, Ward JD. Is ACE inhi- 227. Schäfer S, Linz W, Vollert H, et al. The vasopepti-
bition with lisinopril helpful in diabetic neuropathy? dase inhibitor AVE7688 ameliorates type 2 diabetic
Diabet Med. 1995;12:307–9. nephropathy. Diabetologia. 2004;47:98–103.
214. Ebihara F, Di Marco GS, Juliano MA, Casarini 228. Spenny ML, Muangman P, Sullivan SR, et  al.
DE.  Neutral endopeptidase expression in mesan- Neutral endopeptidase inhibition in diabetic wound
gial cells. J Renin-Angiotensin-Aldosterone Syst. repair. Wound Repair Regen. 2002;10:295–301.
2003;4:228–33. 229. Wihler C, Schafer S, Schmid K, et  al. Renal accu-
215. Muangman P, Spenny ML, Tamura RN, Gibran mulation and clearance of advanced glycation end-­
NS. Fatty acids and glucose increase neutral endo- products in type 2 diabetic nephropathy: effect of
276 M. Yorek

angiotensin-converting enzyme and vasopeptidase 244. Shaikh SR, Kinnun JJ, Leng X, Williams JA, Wassall
inhibition. Diabetologia. 2005;48:1645–53. SR. How polyunsaturated fatty acids modify molec-
230. Oltman CL, Davidson EP, Coppey LJ, Kleinschmidt ular organization in membranes: insight from NMR
TL, Dake B, Yorek MA. Role of the effect of inhibi- studies of model systems. Biochim Biophys Acta.
tion of neutral endopeptidase on vascular and neural 2015;1848:211–9.
complications in streptozotocin-induced diabetic 245. Simopoulos AP.  The importance of the omega-6/
rats. Eur J Pharmacol. 2001;650:556–62. omega-3 fatty acid ratio in cardiovascular dis-
231. Coppey L, Davidson E, Lu B, Gerard C, Yorek ease and other chronic diseases. Exp Biol Med.
M.  Vasopeptidase inhibitor ilepatril (AVE7688) 2008;233:674–88.
prevents obesity- and diabetes-induced neu- 246. Gil A, Serra-Majem L, Calder PC, Uauy
ropathy in C57Bl/6J mice. Neuropharmacology. R. Systematic reviews of the role of omega-3 fatty
2011;60:259–66. acids in the prevention and treatment of disease. Br J
232. Davidson EP, Kleinschmidt TL, Oltman CL, Lund Nutr. 2012;107:S1–2.
DD, Yorek MA.  Treatment of streptozotocin- 247. Kaliannan K, Li X-Y, Wang B, et  al. Multi-omic
induced diabetic rats with AVE7688, a vasopepti- analysis in transgenic mice implicates omega-6/
dase inhibitor: effect on vascular and neural disease. omega-3 fatty acid imbalance as a risk factor for
Diabetes. 2007;56:355–62. chronic disease. Commun Biol. 2019;2:276.
233. Oltman CL, Davidson EP, Coppey LJ, Kleinschmidt 248. Lanchais K, Capel F, Tournadre A. Could omega-3
TL, Yorek MA.  Treatment of Zucker diabetic fatty fatty acids preserve muscle health in rheumatoid
rats with AVE7688 improves vascular and neural arthritis? Nutrients. 2020;12:E223.
dysfunction. Diabetes Obes Metab. 2009;11:223–33. 249. Ueno Y, Miyamoto N, Yamashiro K, Tanaka R,
234. Davidson EP, Coppey LJ, Yorek MA. Early loss of Hattori N. Omega-3 polyunsaturated fatty acids and
innervation of cornea epithelium in streptozotocin-­ stroke burden. Int J Mol Sci. 2019;20:E5549.
induced type 1 diabetic rats: improvement with 250. Li X, Bi X, Wang S, Zhang Z, Li F, Zhao
ilepatril treatment. Invest Ophthalmol Vis Sci. AZ.  Therapeutic potential of ω-3 polyunsaturated
2012;53:8067–74. fatty acids in human autoimmune diseases. Front
235. Yorek MS, Obrosov A, Lu B, Gerard C, Kardon Immunol. 2019;10:2241.
RH, Yorek MA.  Effect of inhibition or deletion of 251. Marton LT, Goulart RA, Carvalho ACA,
neutral endopeptidase on neuropathic endpoints in Barbalho SM.  Omega fatty acids and inflamma-
high fat fed/low dose streptozotocin-treated mice. J tory bowel diseases: an overview. Int J Mol Sci.
Neuropathol Exp Neurol. 2016;75:1072–80. 2019;20:E4851.
236. von Lueder TG, Atar D, Krum H.  Current role of 252. McGlory C, Calder PC, Nunes EA.  The influence
neprilysin inhibitors in hypertension and heart fail- of omega-3 fatty acids on skeletal muscle protein
ure. Pharmacol Ther. 2014;144:41–9. turnover in health, disuse, and disease. Front Nutr.
237. Sangaralingham LR, Sangaralingham SJ, Shah ND, 2019;6:144.
Yao X, Dunlay SM. Adoption of sacubitril/valsartan 253. Avallone R, Vitale G, Bertolotti M.  Omega-3 fatty
for the management of patients with heart failure. acids and neurodegenerative diseases: new evidence
Circ Heart Fail. 2018;11:e004302. in clinical trials. Int J Mol Sci. 2019;20:E4256.
238. Davidson EP, Coppey LJ, Shevalye H, Obrosov 254. Abdolmaleki F, Kovanen PT, Mardani R, Gheibi-­
A, Yorek MA.  Vascular and neural complications Hayat SM, Bo S, Sahebkar A. Resolvins: emerging
in type 2 diabetic rats: improvement by sacubitril/ players in autoimmune and inflammatory diseases.
valsartan greater than valsartan alone. Diabetes. Clin Rev Allergy Immunol. 2020;58:82–91.
2018;67:1616–26. 255. Shevalye H, Yorek MS, Coppey LJ, et  al. Effect
239. Serhan CN, Chiang N, Dalli J.  New pro-­resolving of enriching the diet with menhaden oil or daily
n-3 mediators bridge resolution of infectious treatment with resolvin D1 on neuropathy in a
inflammation to tissue regeneration. Mol Asp Med. mouse model of type 2 diabetes. J Neurophysiol.
2018;64:1–17. 2015;114:199–208.
240. Serhan CN, Yacoubian S, Yang R. Anti-inflammatory 256. Coppey LJ, Davidson EP, Obrosov A, Yorek
and proresolving lipid mediators. Annu Rev Pathol. MA. Enriching the diet with menhaden oil improves
2008;3:279–312. peripheral neuropathy in streptozotocin-induced type
241. Grisold A, Callaghan BC, Feldman EL.  Mediators 1 diabetic rats. J Neurophysiol. 2015;113:701–8.
of diabetic neuropathy: is hyperglycemia the only 257. Coppey LJ, Holmes A, Davidson EP, Yorek
culprit? Curr Opin Endocrinol Diabetes Obes. MA.  Partial replacement with menhaden oil
2017;24:103–11. improves peripheral neuropathy in high fat fed low
242. Bittner V.  Implications for REDUCE IT in clinical dose streptozotocin type 2 diabetic rat. J Nutr Metab.
practice. Prog Cardiovasc Dis. 2019;62:395–400. 2012;2012:950517.
243. Kris-Etherton PM, Richter CK, Bowen KJ, et  al. 258. Obrosov A, Coppey LJ, Shevalye H, Yorek
Recent clinical trials shed new light on the cardio- MA.  Effect of fish oil vs. resolvin D1, E1, methyl
vascular benefits of omega-3 fatty acids. Methodist esters of resolvins D1 or D2 on diabetic peripheral
Debakey Cardiovasc J. 2019;15:171–8. neuropathy. J Neurol Neurophysiol. 2017;8(453)
Metabolic Mechanisms in Diabetic Neuropathy 277

259. Yorek MS, Coppey LJ, Shevalye H, Obrosov A, endpoints in high-fat-fed/low-dose streptozotocin-­
Kardon RH, Yorek MA. Effect of treatment with sal- treated Sprague Dawley rats. J Diabetes Res.
salate, menhaden oil, combination of salsalate and 2018;2018:2967127.
menhaden oil, or resolvin D1 of C57Bl/6J type 1 dia- 263. Superko HR, Superko SM, Nasir K, Agatston
betic mouse on neuropathic endpoints. J Nutr Metab. A, Garrett BC.  Omega-3 fatty acid blood levels.
2016;2016:5905891. Circulation. 2013;128:2154–61.
260. Goldfine AB, Fonseca V, Jablonski KA, et  al. 264. Laidlaw M, Holub BJ.  Effects of supplemen-
Salicylate (salsalate) in patients with type 2 diabetes: tation with fish oil-derived n-3 fatty acids and
a randomized trial. Ann Intern Med. 2013;159:1–12. alpha-­linolenic acid on circulating plasma lipids
261. Goldfine AB, Fonseca V, Jablonski KA, et  al. The and fatty acid profiles in women. Am J Clin Nutr.
effects of salsalate on glycemic control in patients 2003;77:37–42.
with type 2 diabetes: a randomized trial. Ann Intern 265. Lewis EJH, Perkins BA, Lovblom LE, Bazinet RP,
Med. 2010;152:346–57. Wolever TMS, Bril V. Effect of omega-3 supplemen-
262. Davidson EP, Coppey LJ, Shevalye H, Obrosov tation on neuropathy in type 1 diabetes: a 12-month
A, Yorek MA.  Effect of dietary content of menha- pilot trial. Neurology. 2017;88:2294–301.
den oil with or without salsalate on neuropathic
Mechanisms of Nerve Injury
in Diabetes: Dyslipidemia,
Bioenergetics, and Oxidative
Damage

Stephanie A. Eid, Mohamed Noureldein,
Masha G. Savelieff, and Eva L. Feldman

1 Introduction PN lacks effective treatments; currently, stan-


dard management consists of glucose control.
Peripheral neuropathy (PN) is a common compli- Systematic analysis of randomized, controlled
cation in patients with prediabetes and diabetes. clinical studies indicate, however, that well-­
The most common PN is a distal symmetric poly- controlled glucose more effectively prevents PN
neuropathy, which occurs in a distal-to-proximal development in type 1 (T1D) versus type 2 (T2D)
pattern of nerve damage, starting in the feet diabetes patients [1]. Several clinical studies have
(Fig.  1a). Patients experience either positive since evaluated additional PN risk factors, which
symptoms, e.g., prickling, tingling, or negative have centered on the metabolic syndrome, and its
symptoms, e.g., numbness. When PN has reached components, obesity and dyslipidemia. Several
mid-calf, patients begin to experience symptoms studies in diverse population have found the meta-
in the fingers (Fig.  1b). While there are other bolic syndrome to be a PN risk factor, independent
types of PN in this patient population, particu- of glycemic status (Fig. 1a, Table 1), in the United
larly autonomic neuropathy, this chapter will States [2–8], China [9–11], Denmark [12, 13],
focus on distal symmetric polyneuropathy, Germany [14, 15], and the Netherlands [16].
referred to as PN. Furthermore, PN risk increases with the number of
metabolic syndrome components [9, 11, 17, 18].
Stephanie A.  Eid, Mohamed Noureldein and Masha Although hyperglycemia remains a major risk
G.  Savelieff contributed equally with all other factor for persons with T2D, the emergence of
contributors.
the metabolic syndrome, especially dyslipidemia
and obesity as additional PN risk factors, suggest
a distinct or additional pathophysiology underly-
S. A. Eid · M. Noureldein
Department of Neurology, University of Michigan ing neuropathy in T2D versus T1D. This is fur-
Medical School, Ann Arbor, MI, USA ther supported by multiple studies showing that
M. G. Savelieff good or intensive control does not necessarily
NeuroNetwork for Emerging Therapies, University of prevent PN development in T2D [1, 19, 20]. In
Michigan, Ann Arbor, MI, USA addition to impaired glucose handling, persons
E. L. Feldman (*) with T2D frequently have comorbid obesity and
Department of Neurology, University of Michigan dyslipidemia [3, 21], suggesting that, even if dia-
Medical School, Ann Arbor, MI, USA betes is a major PN driver, the effects of lipids on
NeuroNetwork for Emerging Therapies, University of nerve health also warrant investigation to fully
Michigan, Ann Arbor, MI, USA appreciate the mechanisms of neurodegeneration
e-mail: efeldman@umich.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 279
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_16
280 S. A. Eid et al.

a b c

Fig. 1  The peripheral nervous system, PN, and sensory fascicle, which houses unmyelinated neurons (C fibers in
neurons. (a) Outline of lean (silhouette on the right) and Remak bundles) and myelinated neurons (A fibers). An
obese (silhouette on the left) body. PN starts in the feet endoneurium covers neuron fibers. Close-up examination
(deep red) and progresses to the calves (fading red). At of a myelinated sensory neuron (magnifying glass) reveals
this point, PN commences in the finger tips (deep red) and the myelinating Schwann cells, supportive glia, which
progresses to the hands (fading red). Schematic lumbar wrap around the neuron axon. The neuronal cell body
spinal cord section is shown with the dorsal root ganglion (otherwise called the soma) resides with the DRG and the
(DRG), which houses sensory neuron cell bodies. dendrites forge connections with spinal cord neurons. Not
Simplified representation of peripheral sciatic nerves shown are the neuron’s axon termini, which reside within
shown, which runs dorsally. The nerve actually branches the target organ, e.g., skin sensory receptors. Red inset:
in the lower leg. (b) Anatomy of a sensory peripheral Neuron cross-section showing central axon and wrapped
nerve. Nerves are structured into bundles of neurons Schwann cell. (c) Schematic of a peripheral sensory nerve
embedded in concentric layers of connective tissue (epi- to illustrate the aspect ratio. For the longest nerves, e.g.,
neurium, perineurium, endoneurium). The epineurium sciatic, the axon is around 20,000 times longer than the
covers the entire nerve structure and holds within it fasci- cell body diameter. Schwann cells only shown in the start
cles, blood vessels (arterial, red; venous, blue), and addi- of the figure, but occur along the entire neuron axon, till
tional loose connective tissue. A perineurium covers each the axon terminus. Created with BioRender.com

in PN. However, prior research focused solely on [22]. Feeding a high-fat diet (HFD) rich in
the effects of hyperglycemia on peripheral ner- ­saturated fatty acids to wild-type mice induces
vous system function, particularly of constituent ­obesity and impaired glucose tolerance resem-
neurons and Schwann cells as isolated systems, bling prediabetes. Additional administration of
which only reveals a fraction of the complete pic- low-­dose streptozotocin (STZ) abrogates insulin
ture. Newer studies addressing the role of dyslip- production and generates a T2D mouse model.
idemia and other metabolic syndrome These diet-based models are devoid of genetic
components and whole nerve bioenergetics are mutations, like the leptin signaling deficient
identifying novel aspects of PN pathogenesis in db/db and ob/ob mice, and instead maintain
T2D. leptin production [23, 24], which is more akin to
These advancements have rested on the devel- hyperleptinemia in obesity [25]. The obese HFD
opment of animal models, which better mirror prediabetes and HFD-STZ T2D mice develop
diet-induced metabolic dysfunction in humans small fiber PN, through intraepidermal nerve
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 281

Table 1  Summary of clinical studies, which identified components of the metabolic syndrome, including obesity and
dyslipidemia, as PN risk factors
Study Country Population Main MetS and diabetes findings related to PN
Callaghan United States The Diabetes Study of Northern TG level was an independent, stepwise risk
et al. (2011) California (DISTANCE) of factor for non-traumatic lower-extremity
28,701 participants with T1D amputations; TG 150–199 mg/dl, HR 1.10
(~3.5%) or T2D (~96.5%) with (95%CI 0.92–1.32); 200–499 mg/dl, 1.27
baseline TG (1.10–1.47); >500 mg/dl, 1.65 (1.30–2.10);
reference <150 mg/dl
Smith et al. United States The Utah Diabetic Neuropathy Multivariate analysis found obesity and TG
(2013) Study (UDNS) of 218 T2D were related to IENFD loss while elevated
participants HbA1c was related to motor NCV defects
Callaghan United States University of Michigan Weight Diabetes OR 4.90 (95%CI 1.06–22.63) and
et al. (2016) Management Program of 102 WC OR 1.24 (95%CI 1.00–1.55) but not
obese participants, 44.1% prediabetes OR 3.82 (95%CI 0.95–15.41)
normoglycemic, 30.4% significantly associated with PN in
prediabetic, 25.5% T2D multivariable models. PN prevalence was high
in this obese population, even in
normoglycemic individuals
Callaghan United States Health, Aging, and Body Diabetes OR 1.65 (95%CI 1.18–2.31) and
et al. (2016) Composition (Health ABC) HbA1c OR 1.42 (95%CI 1.15–1.75) were the
study of 2382 participants, only Metabolic syndrome measures
21.0% diabetic, 29.9% significantly associated with PN. WC and
prediabetic, 52.8% metabolic HDL significantly associated with multiple
syndrome secondary PN outcomes
Jaiswal et al. United States 79 T2D Pima Indian Participants with versus without PN were more
(2016) participants obese (BMI 36 ± 9 vs 32 ± 5 kg/m2, p = 0.027)
Jaiswal et al. United States SEARCH for Diabetes in Youth Of Metabolic syndrome components, higher
(2017) study of 1734 T1D youths and LDL-c, TG, obesity, diastolic BP and lower
258 T2D youths HDL-c were PN risk factors in T1D youths;
lower HDL-c was a PN risk factor in T2D
youths. Poor glycemic control correlated with
PN in T1D OR 1.53 (95%CI 1.24–1.88) but
not T2D youths OR 1.05 (95%CI 0.7–1.56)
Callaghan United States University of Michigan bariatric BMI was comparable in obese participants
et al. (2020) surgery clinic of 138 obese with and without PN (p = 0.86). WC OR 1.39
participants (95%CI 1.10–1.75), TGs OR 1.31 (95%CI
1.00–1.70), systolic BP OR 2.89 (95%CI
1.49–5.61) were significantly associated with
PN
Lu et al. China Shanghai Diabetic Neuropathy Participants with known T2D had the highest
(2013) Epidemiology and Molecular PN frequency (13.1%). Among participants
Genetics Study (SH-DREAMS) without T2D, higher WC, 2-h postprandial
of 2035 participants, 22.5% plasma glucose, and hypertension were more
normoglycemic, 51.3% likely in those with PN
prediabetic, 26.2% T2D
Han et al. China Shanghai Diabetic Neuropathy Increase Metabolic syndrome in components
(2015) Epidemiology and Molecular linearly proportional to PN (p = 0.001 for
Genetics Study (SH-DREAMS) trend). Metabolic syndrome OR 2.0 (95%CI
of 2035 participants 1.2–3.2) for PN; however, Metabolic syndrome
no longer associated with PN when HOMA-IR
was added to the binary logistic regression,
HOMA-IR OR 1.2 (95%CI 1.1–1.4) associated
with PN
Callaghan China Pinggu, China study of 4002 Diabetes OR 2.60 (95%CI 1.77–3.80), weight
et al. (2018) participants, 37.2% OR 1.09 (95%CI 1.02–1.18), and the number
normoglycemic, 18.9% of metabolic syndrome components OR 1.17
diabetic, 44.0% prediabetic (95%CI 1.03–1.32) were significantly
associated with PN
(continued)
282 S. A. Eid et al.

Table 1 (continued)
Study Country Population Main MetS and diabetes findings related to PN
Andersen Denmark Danish arm of the Anglo- Baseline weight HR 1.09 (95%CI 1.03–1.16),
et al. (2018) Danish-Dutch study of WC HR 1.14 (95%CI 1.05–1.24), BMI HR
Intensive Treatment of Diabetes 1.14 (95%CI 1.06–1.23), log2(methylglyoxal)
in Primary Care (ADDITION) HR 1.45 (95%CI 1.12–1.89), HDL-c HR 0.82
of 1256 T2D participants (95%CI 0.69–0.99), and LDL-c HR 0.92
without PN at baseline (95%CI 0.86–0.98) significantly associated
with incident PN
Christensen Denmark Danish Centre for Strategic In regression analyses, central obesity (WC,
et al. (2020) Research in Type 2 Diabetes WHR, waist-to-­height ratio) associated with
(DD2) Cohort of 5249 T2D PN. Of Metabolic syndrome components, TG
participants ≥1.7 mmol/l aPR 1.36 (95%CI 1.17–1.59),
HDL-c <1.0/1.2 mmol/l (male/female) aPR
1.35 (95%CI 1.12–1.62), hs-CRP ≥3.0 mg/l
aPR 1.66 (95%CI 1.42–1.94), and HbA1c ≥78
mmol/mol (9.3%) aPR 1.42 (95%CI 1.06–
1.88) associated with PN
Ziegler et al. Germany MONICA (Monitoring Trends WC and diabetes were independent factors
(2008) and Determinants on significantly associated with PN in the entire
Cardiovascular Diseases)/ population. Peripheral arterial disease was
KORA (Cooperative Research additionally associated with PN in the diabetic
in the Region of Augsburg) of population
195 diabetes, 46 IGT, 71 IFG,
81 NGT participants
Schlesinger Germany Cooperative Health Research in Overweight OR 3.06 (95%CI 1.57–5.97),
et al. (2019) the Region of Augsburg obese OR 3.47 (95%CI 1.72–7.00), WC OR
(KORA) F4/FF4 cohort of 513 1.22 (95%CI 1.07–1.38). Interaction analyses
participants did not reveal any differences by diabetes
status
Hanewinckel Netherlands Rotterdam Study of 908 BMI independently associated with lower sural
et al. (2017) participants SNAP (OR 1.53 95%CI 1.13–2.09) and
peroneal CMAP (OR 1.49 95%CI 1.11–1.99)
amplitudes
aPR adjusted prevalence ratio, BMI body mass index, BP blood pressure, CI confidence interval, CMAP compound
motor action potential, HbA1c hemoglobin A1c, HDL-c high-­density lipoprotein cholesterol, HOMA-IR homeostatic
model assessment of insulin resistance, HR hazard ratio, hs-CRP high-sensitivity C-reactive protein, IENFD intraepi-
dermal nerve fiber density, IFG impaired fasting glucose, IGT impaired glucose tolerance, LDL-c low-density lipopro-
tein cholesterol, Metabolic syndrome metabolic syndrome, NCV nerve conduction velocity, NGT normal glucose
tolerance, OR odds ratio, SNAP sural sensory nerve action potential, T2D type 2 diabetes, TG triglycerides, WC waist
circumference, WHR waist-to-hip ratio

fiber density loss, and large fiber PN, with lights new therapeutic avenues. Although evi-
decreased nerve conduction velocities in both dence from large, randomized, and controlled
sensory sural and motor sciatic nerves [26–29]. clinical trials remains scarce, the emerging body
Furthermore, omics and integrated multi-omics of work which suggests lifestyle changes to cor-
platforms are providing insight through unbiased rect the metabolic syndrome and promote weight
interrogation of dysregulated pathways in neuro- loss could prevent PN onset or slow progression.
pathic nerve from animal models as well as The American Diabetes Association now recom-
human biopsy samples. These studies are uncov- mends diet and exercise as the first-line PN treat-
ering participation of multiple pathways includ- ment to reverse metabolic syndrome components
ing inflammation and oxidative stress in PN and improve glucose handling and lipid profiles
pathophysiology. [30–34].
Lastly, the association of metabolic syndrome This chapter will examine these three emer-
and its components with PN development high- gent facets of PN pathophysiology. First, we will
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 283

examine bioenergetics in the diabetic nerve and transported into neurons, glucose catabolism
the effects of nutrient overload, followed by a commences anaerobically within the cytoplasm
discussion of bioenergetics with a lipid-centric by glycolysis, which releases two ATP molecules
focus to lay out the effects of dyslipidemia and and pyruvate [38]. The pyruvate is shuttled into
altered nerve lipid metabolism on PN develop- the mitochondrial matrix by mitochondrial pyru-
ment. Finally, this chapter will round off with the vate carrier [43], where it is further metabolized
most promising therapeutic avenues for PN based through the tricarboxylic acid cycle, generating
on the lipid-centric perspective, particularly for guanosine triphosphate (GTP), reduced nicotin-
T2D patients that also have the metabolic amide adenine dinucleotide (NADH), and flavin
syndrome. adenine dinucleotide (FADH2; Fig. 2c) [44, 45].
Oxidative phosphorylation next utilizes NADH
and FADH2 as cofactors to complexes I and II,
2 Bioenergetics and Nutrient respectively, which are electron transport chain
Overload in the Nerve proteins that reside within the inner mitochon-
and PN drial membrane [46, 47]. Electrons continue to
move along the electron transport chain, on to
2.1 Introduction to Nerve complexes III and IV [48, 49], while protons are
Bioenergetics transported from the matrix to the intermembrane
space [45]. This transport generates a proton gra-
Peripheral neuron morphology and its high dient and the mitochondrial membrane potential
energy demands for signal transmission present a across the inner mitochondrial membrane. The
unique challenge to the peripheral nervous sys- mitochondrial membrane potential is then dissi-
tem. Structurally, peripheral nerve axons can be pated as protons move though complex V (ATP
as long as 3 feet and are 20,000 times the size of synthase), generating ATP [50]. In sum, a single
their cell body (Fig. 1b); thus, energy utilization glucose molecule optimally releases 36 ATP after
and production must be coordinated over propor- tricarboxylic acid cycle and oxidative
tionately large distances. Functionally, peripheral phosphorylation.
neurons require massive amounts of energy to Mitochondrial aerobic oxidation releases
maintain their membrane potential across a large more energy per glucose molecule than anaero-
surface area [35] and high energy density at spe- bic glycolysis; however, It remains uncertain
cialized structures, e.g., at terminal synapses, to what pathway neurons prefer under basal con-
perform their roles [36, 37]. Thus, neurons need ditions, although periods of high-intensity
to orchestrate energy production according to activity may require both [38, 51, 52]. Glucose
their distinct morphology and at areas of highest is believed to be the preferred energy source;
energy consumption [38]. Even under resting however, nerves can develop insulin resistance
conditions, a single neuron can consume 4.7 bil- [53–55], hindering their capacity for glucose
lion adenosine triphosphate (ATP) molecules per uptake.
second [39]. To meet this immense energy Fatty acids are another fuel substrate; they are
demand, neurons rely on efficient bioenergetics catabolized in mitochondria by β-oxidation
through multiple pathways, which is essential to (Fig.  2c) [56]. The fatty acid is first ligated to
their health and survival and for peripheral ner- CoA by acyl-CoA synthetase long-chain family
vous system functioning. member 1 (ACSL1) to generate fatty acyl-CoA,
To generate this energy, sensory neurons and which is shuttled across the outer mitochondrial
axons can utilize glucose and fatty acids, membrane by carnitine palmitoyltransferase I
(Fig. 2a). Glucose uptake is facilitated by glucose (CPT1) that in turn substitutes the CoA for carni-
transporters (GLUTs), which exist in different tine. The acylcarnitine is then translocated across
isoforms (Fig. 2b). Peripheral neurons primarily the inner mitochondrial membrane by CPT2,
express insulin-independent GLUT3 [42]. Once which substitutes the carnitine back to CoA,
284 S. A. Eid et al.

b
c

Fig. 2  Homeostatic peripheral nerve bioenergetics. (a) axon terminals. (c) PYR is transported by mitochondrial
Anatomy of a myelinated sensory neuron. Sensory neu- pyruvate carrier (MPC) across the inner mitochondrial
rons under basal and active conditions require constant membrane (IMM) into the mitochondrial matrix. The
ATP to meet their energy demands. (b) Neurons mainly pyruvate dehydrogenase complex converts PYR into
express insulin-independent glucose transporter (GLUT) acetyl-­CoA, which enters the TCA cycle, generating gua-
3 as well as GLUT1 to transport glucose (GLU) into nosine triphosphate (GTP), reduced nicotinamide adenine
axons as fuel substrate under homeostatic conditions. dinucleotide (NADH), and flavin adenine dinucleotide
GLU is catabolized to pyruvate (PYR) by glycolysis in the (FADH2). NADH generated from the TCA cycle drives
axon cytoplasm, releasing two ATP molecules. oxidative phosphorylation (OXPHOS) within the IMM
Alternatively, neurons do express fatty acid (FA) trans- and intermembrane space, releasing additional ATP mol-
porters (FAT) [40] and may metabolize fatty acids. Under ecules. Alternatively, pyruvate carboxylase converts PYR
homeostatic conditions, Schwann cells utilize GLU, into oxaloacetate, an anaplerotic reaction, which boosts
whereas FAs are used mainly to maintain myelin, although TCA capacity. FAs are ligated to CoA by ACSL1 (acyl-­
they can also be metabolized [41]. Under conditions of CoA synthetase long chain family member 1) to generate
especially high neuronal energy demands, Schwann cells fatty acyl-CoA, which is shuttled across the outer mito-
may supplement neuronal energy reserves via axo-glial chondrial membrane (OMM) by carnitine palmitoyltrans-
crosstalk. PYR is converted to lactate (LAC), which is ferase I (CPT1), which substitutes the CoA for carnitine.
shuttled to neurons through monocarboxylate transporters The acylcarnitine is then translocated across the IMM by
(MCT). Within neurons, LAC is reverted to PYR, which CPT2, which reverts the carnitine back to CoA, releasing
joins the pool of substrates entering the tricarboxylic acid the fatty acyl-CoA into the mitochondrial matrix to enter
(TCA) cycle. Vital mitochondria (colored red) are traf- β-oxidation. Created with BioRender.com
ficked along axons to meet local energy demands, e.g., at

releasing the fatty acyl-CoA into the mitochon- acyl-CoAs go through the same process, except
drial matrix where it undergoes β-oxidation. that the final cleavage yields propionyl-CoA in
Even-chained acyl-CoAs undergo a repeated addition to acetyl-CoA. Subsequently, the gener-
four-step process, which cleaves off two carbon ated acetyl-CoA feeds into the tricarboxylic acid
atoms at a time in the form of acetyl-CoA, as well cycle followed by oxidative phosphorylation to
as NADH and FADH2 [56, 57]. Odd-chained generate ATP.
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 285

2.2 Axo-glial Metabolic The impact of MCT1 knockout may be modest


Interactions in the Nerve under baseline conditions, but aggravated in neu-
ropathic conditions. For instance, Schwann cell-­
Schwann cells are the primary glia in the periph- specific heterozygous MCT1 knockout in mice
eral nerves, which support and nurture neurons. with STZ-induced T1D worsens PN, assessed by
They wrap around axons forming the myelin tail nerve conduction velocity [72]. Complete
sheath (Fig. 2a) that aids in neuronal signal trans- Schwann cell-specific MCT1 deletion in mice
duction. Schwann cells are thought to primarily preferentially impairs sensory over motor nerve
utilize glucose as an energy source to conserve function, but also generates a hypomyelinated
fatty acids for myelin synthesis [41, 58], although phenotype [73]. Furthermore, MCT1 deletion
this remains incompletely known. Indeed, under disrupts lipid synthesis and glycolysis in Schwann
normal physiologic conditions, Schwann cells cells, which revert to a less mature state.
express GLUT1, which may be insulin responsive Overall, evidence indicates an intimate bioen-
[59], and GLUT3 (Fig.  2b) [60, 61]. Thus, they ergetics cooperation between axon function and
undergo insulin-sensitive, facilitated glucose Schwann cells, suggesting prediabetic and dia-
transport via GLUT1 [62] and insulin-­independent betic PN progression may be noncell autono-
glucose transport via GLUT3 [63]. In addition to mous. Thus, metabolic insults that are injurious
their myelinating role, Schwann cells can also to Schwann cells may also contribute to PN.
sense axonal activity and supplement axons with
energy substrates, such as lactate, during periods
of intense neuronal activity [64, 65] via monocar- 2.3 Mitochondrial Dynamics
boxylate transporters (MCTs). in the Nerve and PN
Numerous model systems have demonstrated
this communication channel of Schwann cell-­ In order for axons to function properly, they
axon crosstalk through MCTs [60, 66, 67], which require an extensive amount of energy, which
are currently believed to be involved in axo-glial they obtain from mitochondria, the “power-
metabolic coupling in the peripheral nervous sys- house” of the cell. As a result, well-coordinated
tem [68]. In an axotomy model of peripheral mitochondrial dynamics are essential to neurons,
nerve injury, Schwann cells stabilize neurons and and comprise biogenesis (fission/fusion bal-
slow axon degeneration post injury by increasing ance), trafficking, and mitophagy. Mitochondrial
glycolysis and transferring lactate to axons via biogenesis occurs primarily in the cell body, fol-
MCTs [69]. Schwann cell-restricted knockout of lowed by trafficking of newly generated mito-
glycolytic enzymes or GLUT1 renders Schwann chondria to locations of high energy demand
cells incapable of lactate production and blocks (Fig. 2b) [74]. This transport can occur over very
their protective effect in axotomized nerves. This long distances, as is the case for the sciatic nerve
occurs without affecting nerve myelination, in humans, which extends over 3 feet. Once
indicative specifically of Schwann cell-axon met- mitochondria arrive at their destination, e.g.,
abolic crosstalk. MCT1 is widely expressed by nodes of Ranvier, growth cones, axonal branch-
Schwann cells, but not by sciatic nerve axons ing points, or synaptic terminals, they start to
[70]. Sciatic nerve axon crush in mice with het- participate in  local energy production through
erozygous MCT1 knockout slows nerve regener- oxidative phosphorylation [75–78]. Additionally,
ation and recovery of neuronal compound motor local mitochondrial biogenesis can occur, e.g., in
action potentials, indicating MCT1 is important neurites, although the extent this occurs remains
to neuronal function. In neurodevelopmental a point of continued research [79–81]. Terminally
models, Schwann cell-restricted MCT1 ablation damaged mitochondria, generally from exces-
leads to defects in motor end-plate innervation, sive oxidative stress, are eliminated by mitoph-
although it does not impact motor and sensory agy to clear the way for new, functioning
nerve conduction velocities nor myelination [71]. mitochondria [82].
286 S. A. Eid et al.

Since mitochondria are crucial to maintaining sion versus fusion. The former is generally
proper nerve function, mitochondrial dysfunction favored in the presence of severe stress or nutri-
and bioenergetics failure are central to and con- ent excess, the latter usually during nutrient star-
sistently associated with PN pathogenesis vation [82]. For example, hyperglycemia induces
(Fig.  3a) [88–90]. Diabetes and the metabolic mitochondrial fission in dorsal root ganglia in
syndrome affect all aspects of mitochondrial vitro and the dorsal and ventral root axons of
dynamics in neurons. Mitochondrial length and/ db/db T2D mice in vivo [91]. On the other hand,
or size are regulated by the balance between fis- dyslipidemia alters the fission–fusion balance

b
c

Fig. 3 Diabetic peripheral nerve bioenergetics. (a) may be impaired in Schwann cells since they express
Sensory neurons undergo distal-to-proximal axon degen- insulin-dependent GLUTs. Under dyslipidemic condi-
eration due to bioenergetics and mitochondrial trafficking tions, Schwann cells have increased expression of ACSL1
breakdown and buildup of reactive oxygen species (ROS) (acyl-CoA synthetase long-chain family member 1; inset
and lipotoxic species. (b) Under hyperglycemic condi- (c)) and carnitine palmitoyltransferase (CPT1), concomi-
tions, glucose (GLU) accumulates in axons through tant with a boost in production of acylcarnitines, oxidative
insulin-­independent glucose transporters (GLUT). stress, and apoptosis [85]. Mitochondrial dysfunction in
Excessive glucose boosts flux through the polyol and hex- Schwann cells shifts lipid metabolism from lipogenesis to
osamine pathways, increases protein crosslinking through lipolysis with a consequent increase acylcarnitines, which
advanced glycation end products (AGEs) and receptor of can diffuse into axons [86]. Acylcarnitines can also trigger
AGEs (RAGE) activation, and raises diacylglycerol levels a decrease in endoplasmic reticulum calcium stores [87]
and protein kinase C (PKC) activation. Neurons can also and an increase in intracellular calcium [86]. (c) Impaired
utilize fatty acids as a fuel substrate; however, large influx bioenergetics and altered metabolic flux through the tri-
and catabolism of fatty acids under dyslipidemic condi- carboxylic acid (TCA) cycle and β-oxidation increases
tions increases ROS generation and lipotoxic acylcarni- ROS production and lipotoxic intermediates. IMM inner
tine production from mitochondria (inset C). Excessive mitochondrial membrane, LAC lactate, MCT monocar-
fatty acids (FA) also impair mitochondrial trafficking, boxylate transporter, MPC mitochondrial pyruvate carrier,
depolarize mitochondria, and alter their bioenergetics [83, OMM outer mitochondrial membrane. Created with
84], leading to immobile, damaged mitochondria (colored BioRender.com
brown). Under hyperglycemic conditions, GLU uptake
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 287

[83, 84]. Nutrient overload also influences mito- diabetic or the metabolic syndrome conditions,
chondrial trafficking (Fig.  3b). For example, hyperglycemia stimulates mitochondrial oxida-
excess saturated fatty acids, a hallmark of dyslip- tive phosphorylation, leading to reactive oxygen
idemia, impairs mitochondrial axonal transport species overproduction, mitochondrial oxidative
in dorsal root ganglia neurons in vitro [83, 84]. In damage, and compromised intracellular pro-
vivo, HFD induces obesity and prediabetes in cesses (Fig. 3c) [98]. In dorsal root ganglia neu-
mice and alters mitochondrial dynamics, includ- rons, hyperglycemia induces reactive oxygen
ing energy production and trafficking in saphe- species production and oxidative stress, as well
nous nerves [29]. as caspase-dependent apoptosis [99].
Interestingly, Charcot-Marie-Tooth disease, Reactive oxygen species overproduction in
the most prevalent inherited neuropathy [74], is diabetes and the metabolic syndrome addition-
also a length-dependent polyneuropathy and is ally inhibit key mitochondrial enzymes and con-
caused, in a large subset of patients, by mutations tribute further to mitochondrial dysfunction and
to genes regulating mitochondrial dynamics, damage [100]. Reactive oxygen species, such as
including bioenergetics, trafficking, fission, and superoxide and hydrogen peroxide, inhibit a key
fusion. Mitochondrial dysfunction is also present tricarboxylic acid cycle enzyme, aconitase [100,
in chemotherapy-induced peripheral neuropathy 101], which disrupts the tricarboxylic acid cycle.
[92, 93], which manifests in a glove and stocking It also results in α-ketoglutarate dehydrogenase
pattern. Thus mitochondrial dysfunction is a cen- (α-KGDH) overactivation that further truncates
tral theme in not only prediabetic and diabetic PN the tricarboxylic acid cycle [100]. α-KGDH itself
in both mouse models and humans but also in is a source of reactive oxygen species, which fur-
other well-established neuropathic disorders. ther exacerbates mitochondrial oxidative damage
[100].
Fortunately, peripheral nervous system
2.4 Mitochondrial Oxidative Schwann cells possess a high antioxidant capac-
Stress in the Nerve and PN ity and can detoxify reactive oxygen species to a
certain limit, even under conditions of
Mitochondria are a major source of reactive oxy- hyperglycemia-­induced metabolic dysregulation
gen species, such as superoxide anions, which [102]. Schwann cells are extremely resilient to
can be produced unintentionally during oxidative hyperglycemia and only become susceptible to
phosphorylation [94], even under normal basal mitochondrial injury at very high glucose con-
conditions. Cellular antioxidant defense mecha- centrations (150 mM) [103]. Furthermore, the
nisms scavenge these reactive oxygen species to basal activity of antioxidant enzymes in Schwann
maintain cellular redox homeostasis and prevent cells rivals the maximum stimulated activities of
oxidative damage [95, 96]. However, oxidative the same enzymes in dorsal root ganglia neurons
stress occurs when reactive oxygen species pro- [102].
duction outstrips the ability of cells to mount an In summary, neurons and their axons are
antioxidant defense. Reactive oxygen species highly reliant on normal mitochondrial dynamics
production from mitochondrial respiration is and are susceptible to injury if mitochondrial
influenced by the mitochondrial membrane function is disrupted by nutrient overload.
potential. Low mitochondrial membrane ­potential Hyperglycemia and/or dyslipidemia shifts the
slows respiration and reduces reactive oxygen dynamics from a homeostatic to a pathological
species production [97], while high mitochon- state, disrupting the balance in mitochondrial fis-
drial membrane potential boosts respiration and sion/fusion, trafficking, and reactive oxygen spe-
increases reactive oxygen species generation cies production. These aspects are present in both
[98]. The mitochondrial membrane potential is diabetic, type 1 and 2, and prediabetic obesity
related to substrate flux and level of oxidative models, highlighting mitochondrial dysfunction
phosphorylation through mitochondria. Under generally as a PN hallmark.
288 S. A. Eid et al.

2.5 Glucose Excess o­ verexpression worsens outcomes [117].


and Inflammation Disappointingly, pharmacological inhibition of
in the Nerve and PN aldose reductase in 32 clinical trials conducted
during the 1990s failed to improve diabetic PN
As mentioned above, neurons mostly express outcomes [118], which may have arisen from the
insulin-independent GLUT3 [42]; therefore, inability of these inhibitors to reach the periph-
insulin-resistant, hyperglycemic, or impaired eral nervous system [119].
glucose tolerance conditions do not affect glu- Another prominent pathway stemming from
cose transport into neurons, resulting in neuronal axonal glucose accumulation, which leads to
glucose accumulation [104–106]. This glucose nerve injury, is the advanced glycation end prod-
buildup has numerous downstream effects ucts (AGEs) pathway. Glucose buildup in the
(Fig. 4), which ultimately lead to neuronal dam- nerve under hyperglycemic conditions can go
age [107, 108]. through the Maillard reaction by reacting with
One well-known pathway leading to injury is protein amino groups to give rise to Schiff bases
the polyol pathway. Glycolysis commences in the or Amadori products. These rearrangement, gen-
cytoplasm by hexokinase, which phosphorylates erating reactive groups, which stably crosslink
glucose, committing it to glycolysis [109–111]. proteins to generate AGEs in the nerve [36, 120,
However, glucose excess outstrips hexokinase 121]. AGEs cause cellular damage by crosslink-
activity and glycolytic capacity, leading to surplus ing critical proteins, inhibiting their function.
glucose, which is instead converted to sorbitol by They also bind to a specific receptor, RAGE
aldose reductase through the polyol pathway [106]. (receptor for AGEs), which triggers a signaling
The accumulating sorbitol disrupts the osmotic cascade leading to nuclear factor-kappa B (NF-­
balance inside axons, inducing osmotic stress and κB) activation and inflammation, vasoconstric-
an efflux of myo-inositol and taurine in an effort to tion, and loss of neurotrophic support in the
restore osmotic balance [112]. Myo-inositol is peripheral nervous system [122].
essential for Na+/K+ ATPase channel functioning; Excess glucose can also increase flux through
thus, myo-inositol loss disrupts Na+/K+ ATPase the hexosamine pathway. During the first few gly-
function and consequently membrane resting colytic steps, glucose is converted to fructose-­6-­
potential and axon electrophysiology [36]. phosphate, which can either remain within
Additionally, aldose reductase utilizes NADPH as glycolysis or be directed into the hexosamine path-
a cofactor; hence, excessive aldose reductase activ- way. Although the shunt into the hexosamine path-
ity depletes NADPH energy stores, interfering with way normally occurs at a low rate, it dramatically
the synthesis of nitric oxide, an important signaling increases under hyperglycemic conditions [107].
molecule, and glutathione, an antioxidant defense The hexosamine pathway utilizes fructose-­ 6-­
molecule [36]. Antioxidant glutathione deficiency phosphate and glutamine in the first step, ultimately
exacerbates the damaging effects of reactive oxy- producing uridine 5'-diphosphate-N-acetylglucos-
gen species, mediating further cellular damage. amine (UDP-­GlcNac). UDP-GlcNac binds to ser-
This “metabolic flux” due to polyol pathway ine/threonine residues of multiple proteins, a
activation is considered a central pathophysio- process termed O-GlcNAcylation, altering their
logic mechanism of metabolic PN [112]. In addi- function and causing downstream injury. For
tion, aldose reductase activation decreases ATP instance, O-GlcNAcylation of specificity protein 1
production, precipitates neuronal injury [113] (Sp1), a key transcription factor, disrupts cellular
and interferes with energy influx from Schwann homeostasis by activating transforming growth
cells [109, 110, 114]. Further literature supports factor-β (TGF-β) and plasminogen activator inhibi-
the deleterious effects of aldose reductase hyper- tor-1 (PAI-1), resulting in peripheral nerve inflam-
activation and the polyol pathway [115]. In mice mation [123].
with STZ-induced T1D PN, lack of aldose reduc- Finally, another glycolytic intermediate
tase preserves nerve function [116], while involved in neuronal dysfunction in hyperglyce-
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 289

a b

Fig. 4  Hyperglycemia and inflammation in the nerve and pathway: Excess F-6-P can exit glycolysis and react with
PN. (a) Under homeostatic conditions, insulin-­ glutamine, ultimately producing uridine 5′-diphosphate-
independent glucose transporter 1 (GLUT1, green trans- N-­acetylglucosamine (UDP-GlcNac). UDP-GlcNac binds
porter) imports glucose (GLU) into axons. GLU is to serine/threonines altering protein function and causing
phosphorylated by hexokinase (HK) into fructose-6-­ downstream injury, e.g., of key transcription factors (TF),
phosphate (F-6-P), committing it to glycolysis and even- disrupting cellular homeostasis by activating transforming
tual pyruvate (PYR) production. The PYR is transported growth factor-β (TGF-β) and plasminogen activator inhib-
into mitochondria, where it enters the tricarboxylic acid itor-­1 (PAI-1), resulting in peripheral nerve inflammation.
(TCA) cycle followed by oxidative phosphorylation (3) Advanced glycation end products (AGEs) pathway:
(OXPHOS) to generate energy. Mitochondria remain vital Excess GLU can go through the Maillard reaction eventu-
and functioning (red). (b) Under hyperglycemic condi- ally giving rise to AGEs, which stably crosslink proteins,
tions, excess GLU floods neuron axons; the subsequent inhibiting their function. AGEs also bind to a specific
increased catabolic flux through the TCA cycle increases receptor, RAGE (receptor for AGEs), triggering signaling
reactive oxygen species (ROS) production, damaging leading to nuclear factor-kappa B (NF-κB) activation,
mitochondria (brown). GLU accumulation also boosts resulting in inflammation, vasoconstriction, and loss of
flux through multiple pathways. (1) Polyol pathway: neurotrophic support in the peripheral nervous system. (4)
When GLU excess outstrips HK activity and glycolytic Protein kinase C (PKC) activation pathway: The accumu-
capacity, surplus GLU is instead converted to sorbitol lating glycolytic intermediate, dihydroxyacetone phos-
(SOR) by aldose reductase (AR). Accumulating SOR dis- phate (DHAP), is converted to diacylglycerol (DAG),
rupts axon osmotic balance, leading to myo-inositol and which in turn activates protein kinase C (PKC), leading to
taurine efflux. Myo-­inositol is essential for Na+/K+ ATPase nerve dysfunction, through insulin resistance, impaired
channel functioning, disrupting membrane resting poten- Na+/K+ ATPase, hypoxia, TGF-β overexpression, and dis-
tial and axon electrophysiology. Additionally, excessive rupted vascular endothelial growth factor. IMM, inner
AR activity depletes NADPH, interfering with antioxidant mitochondrial membrane; MPC, mitochondrial pyruvate
defense, e.g., glutathione (GSH) synthesis, further exacer- carrier; OMM, outer mitochondrial membrane. Created
bating ROS levels and oxidative stress. (2) Hexosamine with BioRender.com
290 S. A. Eid et al.

mia is dihydroxyacetone phosphate (DHAP). additionally identified elevated triglycerides and


Upon accumulation, DHAP is converted to diac- anthropometric measures waist-to-hip and waist-
ylglycerol, which in turn activates protein kinase to-height ratios as PN risks [13]. We found that
C (PKC) [124]. Activated PKC is responsible for HDL associated with multiple secondary PN out-
multiple metabolic and inflammatory pathways comes in the Health, Aging, and Body Composition
that can result in nerve dysfunction, such as insu- (Health ABC) study [3] and triglycerides were a
lin resistance, impaired Na+/K+ ATPase, hypoxia, stepwise risk factor for nontraumatic lower-
TGF-β overexpression, and vascular endothelial extremity amputations [7]. In a population of
growth factor disruption [125]. Once again, the youths, lower HDL-c was a PN risk factor in T2D
idea of using a PKC inhibitor to treat diabetic PN [5]; notably, however, higher LDL-c and triglycer-
failed in clinical trials [125]. Similar to the clini- ides and lower HDL-c were PN risk factors in T1D
cal narrative for aldose reductase inhibitors, the youths, although this cohort was significantly less
failure of PKC inhibitors was attributed to the obese than the T2D.  Therefore, dyslipidemia in
inability of the drugs to penetrate into the periph- patients with T1D may also have implications for
eral nervous system [126]. PN onset, although this has not been extensively
studied. Patients with T1D historically suffer from
poor weight, but obesity, as in T2D, is similarly
3 Dyslipidemia Is Associated becoming more prevalent in T1D [133, 134].
with PN It is also important to note that obesity is intri-
cately tied to dyslipidemia through fat depots and
Although glycemic control is generally effective energy balance and flux in the body [135, 136]. It
for preventing or slowing PN progression in T1D, is therefore unsurprising that multiple measures
the risk of PN persists in prediabetic and T2D of obesity, such as body mass index and waist cir-
patients, even with well-controlled glycemia cumference, have also correlated with PN risk
[127]. Dyslipidemia, commonly seen clinically in (Table 1). Specifically, central obesity is the most
both prediabetes and T2D patients, has recently significant PN driver [2, 4] compared to other
emerged as an independent PN risk factor [8, 128– anthropometric measures. This is anticipated,
131]. Dyslipidemia refers to a range of plasma since the visceral, intra-abdominal fat depot is
lipid abnormalities, characterized by the triad of more metabolically impaired, hypertrophied, and
increased triglycerides and low-density lipopro- pro-inflammatory, compared to other depots,
tein cholesterol (LDL-c) and decreased high-den- such as gluteal, femoral, and subcutaneous [135,
sity lipoprotein cholesterol (HDL-c) [132]. 137–140]. Thus, an understanding of systemic
Several clinical studies have identified dyslip- energy balance and lipid trafficking between the
idemia, either in sum or through its individual visceral fat depot and circulation is relevant to
components (elevated triglycerides and LDL-c or dyslipidemia and downstream events.
lowered HDL-c), to contribute to PN risk (Table 1). The association of dyslipidemia, as well as
This is perhaps best exemplified by the Danish obesity, with PN is strengthened by preclinical
arm of the Anglo-Danish-Dutch study of Intensive studies in HFD-induced obese mice, which
Treatment of Diabetes in Primary Care develop dyslipidemia and PN independent of
(ADDITION), a longitudinal study of 1256 T2D hyperglycemia [26, 28, 141]. Overall, these find-
participants without PN at baseline, which were ings suggest that the underlying PN pathophysi-
assessed for incident PN at a mean follow-up of 13 ology may differ in T1D versus T2D [127], and
years [12]. Low baseline HDL-c was protective for this increasingly may be the more accurate per-
future PN, whereas higher waist circumference spective that dyslipidemia may add an additional
and body mass index were predictive of future layer of pathological processes over those
PN.  Another study conducted in Denmark, the induced by hyperglycemia alone. In fact, glucose
Danish Centre for Strategic Research in Type 2 and lipid metabolism acts in concert systemically
Diabetes (DD2) Cohort of 5249 T2D participants in the body overall; thus, an understanding of
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 291

systemic metabolic dynamics is needed to impart system, beginning with lipid uptake, lipogenesis,
an understanding in target organs. and lipotoxic species accumulation. Next, within
As a more recent avenue of investigation, the the nerve, we will discuss the cellular and molec-
mechanisms underlying the effects of dyslipid- ular influence of lipids on mitochondrial bioener-
emia and altered lipid metabolism on PN remain getics and trafficking, oxidative stress, and
incompletely understood. This section will inflammation, and their implications on nerve
review several aspects of lipid metabolism, start- health and PN (Fig.  5). Whenever possible, we
ing from global systemic effects of dyslipidemia will also highlight cell-specific behavior, e.g.,
and circulating lipid levels, to the localized neuronal, Schwann cells, and potential lipid-­
impact of dyslipidemia in the peripheral nervous centric axo-glial metabolic crosstalk.

a b

Fig. 5  Lipid metabolism in obesity and dyslipidemia. (a) PN. (3) Lipid accumulation and lipotoxic species: Lipid
Under homeostatic conditions, a balance exists between build-up and upregulated lipid metabolism occurs in neu-
energy production and energy expenditure. Surplus ropathic nerve. High metabolic flux promotes lipid inter-
energy is stored in adipose tissue depots, which act as mediate formation, e.g., ceramides and diacylglycerols.
short- and long-term storage units and as endocrine Schwann cells (SC) overexpress ACSL1 (acyl-CoA syn-
sources. Under excessive caloric intake, visceral fat depot thetase long chain family member 1), which boosts acyl-
hypertrophies and inflames (red inset, immune cell carnitine production. (4) Dyslipidemia induced
recruitment and activation), increasing circulating free inflammation: Saturated fatty acids (SFA) activate toll-­
fatty acids (FA) and pro-inflammatory adipokines. This like receptors (TLR); dyslipidemia increases oxidized
causes insulin resistance, triglyceride release from the low-density lipoproteins (oxLDLs), which activate lectin-­
liver, and ectopic fatty deposition (shown in the fatty like oxLDL receptor (LOX-1), the receptor for oxLDLs;
liver). The changes in circulating lipids leads to dyslipid- these interactions increase inflammation downstream. (5)
emia and the metabolic syndrome. (b) Systemic dyslipid- Dyslipidemia impairs mitochondrial bioenergetics and
emic conditions have localized effects in the nerve (yellow dynamics: Dyslipidemia impairs bioenergetics, biogene-
inset), e.g., lipid uptake (inset (c)). (c) Possible dysregu- sis, trafficking (stop sign over microtubule motor protein),
lated aspects of lipid metabolism in the nerve in PN (blue and reactive oxygen species production, causing mito-
inset). (1) De novo lipogenesis: The LXR-SREBP1-FAS chondrial damage (brown), ultimately leading to PN. FAS,
axis (green) is dysregulated in PN, impairing lipid synthe- fatty acid synthase; LXR, liver X receptor; SREBP1, ste-
sis. (2) Lipid uptake: Increased expression of fatty acid rol regulatory element-binding proteins 1. Created with
transport proteins like CD36 (gray transporter) occurs in BioRender.com
292 S. A. Eid et al.

3.1 Global Energy Imbalance circulating free fatty acids. Thus, dyslipidemia and
and Metabolic Disruption hyperglycemia work in tandem to exacerbate meta-
During Dyslipidemia bolic dysfunction, with consequent dual effects on
the nerve. Two, obesity, e.g., through a hypertro-
Body weight is maintained by a balance between phied visceral depot, elevates free fatty acids and
energy production and energy expenditure. pro-inflammatory cytokines, which can ultimately
Surplus energy substrate from extra food intake affect the nerves and promote PN [135]. Three,
is stored in adipose tissue depots, which act both elevated free fatty acids can be converted by reac-
as short- and long-term storage units and as endo- tive oxygen species to toxic metabolites, such as
crine sources by releasing adipokines [142, 143]. lipid peroxides, leading to systemic inflammation
Adipokines constitute a type of cytokines and and localized neuronal apoptosis [99]. Four, fatty
hormones regulating appetite, insulin sensitivity, acids released from visceral fat depots can accumu-
inflammation, and immune cell recruitment [142, late ectopically in various tissues leading to dam-
144]. During periods of fasting or starvation, adi- age, e.g., muscle, liver as fatty liver disease, as well
pose tissue releases energy in the form of free as in the nerve (Fig. 5b) [28]. Cumulatively, sys-
fatty acids, which are utilized by other organs for temic global energy imbalance can result in a
energy production [143]. This normal physiolog- vicious feed forward cycle of mounting inflamma-
ical process occurs as long as the adipose tissue is tion, insulin resistance, and dyslipidemia, culmi-
healthy and caloric intake is balanced against nating in peripheral nerve injury.
energy expenditure.
In case of excessive caloric intake, adipocytes
enlarge (hypertrophy) and proliferate ­(hyperplasia) 3.2 De Novo Lipogenesis
(Fig.  5a). Particularly, visceral fat depot adipo- in the Nerve and PN
cytes hypertrophy, impairing their ability to store
fat [135, 137–140] and activating adipose triglyc- Systemic energy consumption is critical to our
eride lipase, which increases free fatty acid release understanding of dyslipidemia development; but,
from adipocytes [145]. The rise in circulating free equally important is a thorough understanding of
fatty acids contributes to insulin resistance and energy utilization at the level of the nerve itself.
dyslipidemia along with intracellular fatty acid Lipids are required in the nerve for manifold pur-
accumulation in target tissues or organs [146, poses. Perhaps foremost, Schwann cells utilize
147]. Additionally, the hypertrophied visceral adi- lipids to generate myelin, a structural and insulat-
pose tissue inflames along with resident macro- ing material, which facilitates axon signal trans-
phage activation [139]. This alters adipokine duction [41]. Lipids are also essential components
secretion from the visceral depot, leading to ele- of organelles, e.g., mitochondria, and influence
vated systemic inflammation, another mechanism membrane composition and fluidity [151, 152],
causing insulin resistance [148]. Since insulin which affect nerve conductance [153].
promotes hepatic fatty acid esterification into tri- Additionally, neurons express fatty acid trans-
glycerides and their release into the circulation porters [40] and can utilize fatty acids as fuel
[149], insulin resistance also leads to changes in substrates in β-oxidation to generate energy for
plasma triglycerides, which correlate with T2D nerve function.
risk [150]. Thus, overall, enlarged, hypertrophied Under homeostatic conditions, neurons and
visceral fat depots impair insulin sensitivity and their supporting glia acquire lipids through two
induce dyslipidemia and the metabolic syndrome mechanisms: de novo lipogenesis and/or uptake
by altering the circulating lipid profile [135]. of plasma lipoproteins or fatty acids. There is
We can infer a few critical points from these currently an increased understanding of the
observations. One, the global systemic effects capacity for Schwann cells to synthesize fatty
underscore the close relationship between glucose, acids de novo, which are required for myelina-
e.g., insulin resistance, and lipid metabolism, e.g., tion, a dynamic process with high-lipid demands
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 293

[154, 155]. Schwann cells express fatty acid syn- ides are initially hydrolyzed by lipoprotein lipase
thase (FAS), the key rate-limiting step in de novo (LPL) in endothelial cells, which are widely
lipogenesis, which is essential for the proper found in adipose tissue and skeletal muscle. This
onset and efficiency of myelination [154]. FAS releases non-esterified free fatty acids, which are
activity is regulated by sterol regulatory element-­ taken up either passively by diffusion or facili-
binding proteins 1 (SREBP1) [156], which works tated by transporters, like CD36 and fatty acid
hand-in-hand with the liver X receptors (LXR), transport proteins (FATP) expressed in multiple
an established regulatory pathway of fatty acid tissue types, including peripheral nerves [41].
lipogenesis [157]. LXR has well-characterized Subsequently, acquired fatty acids are either
roles in maintaining fatty acid homeostasis in incorporated into myelin in Schwann cells,
metabolically active tissues [158]. directly oxidized for energy production in mito-
Pathologic conditions dysregulates the process chondria, or stored in lipid droplets [66, 167].
of de novo lipogenesis and the LXR-­SREBP1-­ While lipid uptake for myelin has been exten-
FAS axis in nerves, which is evident from PN sively studied and recently reviewed [41], less is
models (Fig. 5c). A recent study found that inhibit- known about its pathological implications in
ing FAS adversely affected lipid synthesis and PN. The peripheral nervous system is exposed to
axonal growth in neuronal cultures [159]. SREBP1 excess fatty acids in prediabetes and T2D, since
and FAS expression are disrupted in sciatic nerve most patients also have dyslipidemia. Additionally,
from rodents with T1D-induced PN and in cul- early studies suggested that dysregulated LPL
tured Schwann cells exposed to metabolic insults activity may contribute to lipotoxicity in PN [168,
[160, 161]. LXR activation is associated with anti- 169]. Altered CD36 expression is another poten-
inflammatory, anti-­ apoptotic, and antioxidative tial mechanism for increased lipid delivery into
properties in diabetic complications [162, 163]. the nerve, which we have consistently shown is
Gavini et al. showed that LXR is transcriptionally elevated in murine and human PN (Fig. 5c) [28,
active in dorsal root ganglia neurons, and its acti- 170, 171]. Concomitant increases in mitogen acti-
vation reduces saturated fatty acid-induced endo- vated protein kinase (MAPK) signaling in neuro-
plasmic reticulum stress and allodynia in a HFD pathic peripheral nerves from db/db support a role
prediabetic mouse model of PN [164]. These find- for CD36  in mediating a MAPK-dependent
ings are complemented by comparative transcrip- inflammatory response [170]. Peroxisome
tomics profiling in peripheral nerves of mice and proliferator-­activated receptor-gamma coactivator
humans with PN, which demonstrate that the LXR (PGC) 1α, a master regulator of mitochondrial
pathway is dysregulated in PN and conserved biogenesis, potentially controls β-oxidation
across species in T1D and T2D [165]. Importantly, through its actions on the fatty acid transporter
emerging data have demonstrated a role for LXR CD36 [172]. We have shown increased PGC1α
activation in reducing reactive oxygen species [29] and CD36 expression in murine and human
generation and preserving Schwann cell integrity PN [28, 170, 171], suggesting dysregulation of
in T1D PN [166]. Together, these data suggest that lipid uptake and β-oxidation occurs in PN.
a dysregulated LXR-SREBP1-FAS axis, by Overall, lipid accumulation may occur in
impairing crucial lipogenesis, may be involved in nerves resulting from the combination of excess
PN, an area requiring further investigation. circulating fatty acid levels and overexperssion of
uptake transporters, which could, in part, under-
lie PN pathogenesis, a view that requires direct
3.3 Lipid Uptake in the Nerve confirmation in human PN. Furthermore, manip-
and PN ulating key lipogenic targets in sensory neurons
of PN-relevant mouse models is critical to under-
In addition to de novo lipid synthesis, neurons stand the effect of de novo lipogenesis and lipid
and Schwann cells acquire lipids from the circu- uptake on axo-glial interactions, independent of
lation. Under homeostatic conditions, triglycer- myelination.
294 S. A. Eid et al.

3.4 Dyslipidemia, Lipid found that the overall lipid content increased in
Metabolism, and Lipotoxic peripheral nerves of T2D mice, but with few sig-
Species in the Nerve nificant correlations in specific lipid species with
During PN plasma. However, there was an intra-class correla-
tion between plasma and nerve triglycerides [175],
Excess lipid accumulation and metabolic flux which are also associated with PN progression in
increase the likelihood that lipotoxic species the experimental and clinical settings [28, 129].
form, especially when fatty acid uptake outpaces These findings elucidate a complex interaction
β-oxidation [173, 174]. Therefore, lipid accumu- between plasma and nerve lipid levels and reveal
lation in peripheral nerves, as may occur during patterns of altered lipid metabolism, including tri-
obesity and dyslipidemia [28], could lead to lipo- glyceride classes, which could be used as PN bio-
toxic species generation. Preclinical studies in markers in the context of prediabetes and T2D.
PN-relevant culture and mouse models are begin- Lipid build-up directly promotes increased lipid
ning to provide insights into these mechanisms in intermediate levels, such as diacylglycerols [176]
PN, ranging from detecting lipid accumulation, and ceramides [177, 178], which could exert lipo-
overrepresentation of fatty acid metabolism, and toxic downstream effects. Indeed, this is the case in
the generation of lipotoxic species. neuropathic nerve from prediabetic and T2D mice,
Omics technologies are aiding these investiga- which accumulates diacylglycerols [28], and has
tions by interrogating dysregulated pathways in several downstream effects relevant to PN, includ-
neuropathic nerve. For instance, transcriptomics ing PKC activation [125]. Clinical studies of lipo-
profiling of peripheral nerves from db/db T2D toxic species in nerve are lacking in humans, given
mice identified fatty acid metabolism as one of the constraints of tissue accessibility, although cor-
the highly enriched pathways in PN [170]. This relation of lipotoxic plasma species with PN have
result has been consistently replicated in human been performed. Given the relevance of ceramides
sural nerve biopsies and robust HFD-induced pre- to obesity and drawing parallels to hereditary neu-
diabetes and T2D mouse models [28, 165]. ropathies [179], we analyzed plasma
Lipidomics analysis of sciatic nerve from HFD 1-­deoxydihydroceramide levels, which increased
prediabetes and HFD-STZ T2D mice revealed step-wise from lean controls, to obese participants,
abnormal accumulation of saturated fatty acid-­ to obese T2D participants, and were highest in
containing triglycerides, demonstrating that per- obese T2D participants with DN [180]. We also
sistent dyslipidemia increases lipid delivery to the conducted an untargeted metabolomics study of
nerve [28]. Integrating lipidomics with transcrip- plasma from T2D participants to identify species
tomics in neuropathic nerves identified elevated correlating with PN status [181]. Although few
expression of diacylglycerol acyltransferase 2 species differed significantly, an overview analysis
(DGAT2), the enzyme required for the last and of all plasma lipids identified a shift in abundance,
committed step in triglyceride synthesis, a finding chain length, and saturation level of fatty acids and
mirrored in sural nerve biopsies from hyperlipid- numerous complex and lipotoxic species, includ-
emic T2D patients with PN [28]. These adverse ing ceramides and acylcarnitines, by T2D and PN
effects were improved by dietary reversal from status. The influence of complex lipid chain length
saturated fatty acid-rich HFD back to a standard and saturation has also been implicated in diabetic
low-fat diet, underscoring the therapeutic benefit nephropathy [182]. Therefore, moving forward,
of restoring nerve lipid homeostasis in PN. identifying these trends and potential specific spe-
To better understand how systemic dyslipid- cies may unlock therapeutic avenues, e.g., inhibi-
emia affects localized nerve lipid metabolism, we tors of complex lipid biosynthesis or signaling,
conducted an integrated transcriptomic and lipido- such as ceramides, obesity treatments [183, 184],
mic analysis of plasma and peripheral nerves from and potential targets for PN treatment.
db/db T2D mice to identify alterations in shared Growing interest in the non-cell autonomous
and unique lipid species by tissue types [175]. We nature of PN and emergent studies are starting to
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 295

point toward a potential axo-glial communication genes and pathways related to inflammation and
in a lipid-mediated manner [66, 185], including immune system dysregulation in db/db T2D mice
lipotoxic species transfer between Schwann cells [188]. A combined epigenomic-transcriptomic
and axons. This concept is reinforced by our results, analysis of human sural nerves from T2D patients
which demonstrate that Schwann cells increase with progressive versus non-progressive PN
ACSL1 expression in vivo in db/db sciatic nerve identified enrichment of immune system response
and in vitro under dyslipidemic conditions [85]. [189]. Comparing gene expression in neuropathic
This was accompanied by elevated expression of peripheral nerves from db/db T2D mice versus
mitochondrial fatty acid transporters, CPT1a and human also highlights inflammation as a highly
CPT1b, concommitant with a boost in production conserved pathway [165]. In our HFD prediabe-
of acylcarnitines, which may be lipotoxic [186, tes mouse model of PN, transcriptomic analysis
187]. We also found that dyslipidemia impaired of sciatic nerve identified involvement of toll-like
Schwann cells mitochondrial bioenergetics and receptor (TLR) pathways [190]. TLR2 and TLR4
induced oxidative stress and apoptosis in vitro [85]. knockout delayed PN onset in obese prediabetic
In mice, Schwann cell-restricted knockout of mice, implicating inflammation in the early
TFAM (mitochondrial transcription factor A gene), stages of PN.  TLRs are receptors on innate
a transcription factor that regulates mitochondrial immune cells, which sense danger signals, e.g.,
DNA replication and downstream oxidative phos- pathogens, emitting pro-inflammatory signals.
phorylation protein production, does not affect Saturated fatty acids activate TLRs [191–193]
Schwann cell survival, but does induce progressive and constitute an immunometabolic link between
PN [86]. Loss of TFAM disrupts Schwann cell dyslipidemia and inflammation.
lipid metabolism, which shifts from lipogenesis to Additionally, dyslipidemia may indirectly
lipolysis, with a consequent accumulation of affect nerve function by mediating oxidative stress,
medium- and long-chain acylcarnitines in sciatic and altering downstream signaling pathways
nerves. In turn, explants of these nerves release [194]. For example, we previously showed that
multiple long-chain acylcarnitine species into cul- dyslipidemia increases oxidized low-density lipo-
ture media, suggesting these acylcarnitnes could proteins (oxLDLs) in dorsal root ganglia, which in
diffuse from Schwann cells into axons in vivo. turn interact with the lectin-like oxLDL receptor,
Furthermore, treating dorsal root ganglia cultures LOX-1 to induce reactive oxygen species genera-
with the acylcarnitine palmitoyl-carnitine signifi- tion and damage [141]. This mechanism may be
cantly elevates intracellular calcium and induces shared in other complication-prone tissues in dia-
axonal blebbing and degeneration upon long-term betes, such as the retina and kidney [195, 196].
incubation [86]. Overall, abnormal lipid metabo- oxLDLs can also exert their apoptotic effects in
lism and consequent injury of Schwann cells, dis- dorsal root ganglia neurons via TLR4 [197].
rupting normal axo-glial communication, could be Therefore, altered energy metabolism, genera-
damaging to axons and lead to PN. tion of toxic lipid intermediates, inflammation,
mitochondrial dysfunction, and oxidative stress
can act in concert in the dyslipidemic milieu to
3.5 Dyslipidemia-Induced promote PN.
Inflammation in the Nerve
During PN
3.6 Dyslipidemia Impairs
Hypertrophied fat depots increase systemic Mitochondrial Bioenergetics
inflammation by releasing pro-inflammatory adi- and Dynamics in the Nerve
pokines, such as interleukin-6 (IL-6) and tumor During PN
necrosis factor-α (TNFα) [140, 142, 144].
Locally, in neuropathic nerve, our transcriptomic We have outlined how systemic dyslipidemia can
studies have consistently found enrichment of affect localized nerve lipid uptake, lipogenesis,
296 S. A. Eid et al.

and accumulation, with consequent lipotoxic [83]. Dyslipidemia also affects mitochondrial
intermediate generation and inflammation. In a trafficking in sensory dorsal root ganglion axons,
previous section, we also discussed how hyper- decreasing the number of motile mitochondria in
glycemia in diabetes and the metabolic syndrome the presence of palmitate [83, 84]. The effect is
elevate flux through the polyol and hexosamine chain-length-dependent; long-chain saturated
pathways, alter RAGE and PKC activation, fatty acids palmitate or stearate impair mitochon-
increase reactive oxygen species production, and drial transport, whereas medium-chain saturated
produce mitochondrial dysfunction, all key fatty acids laurate and myristate exert no effect
mechanisms proposed for PN pathogenesis [107, [83]. In contrast to saturated fatty acids, unsatu-
108]. With respect to comparing peripheral ner- rated fatty acids are neuroprotective. For exam-
vous system mitochondrial bioenergetics in the ple, the monounsaturated fatty acid oleate
setting of excess glucose and lipids, the db/db prevents palmitate-induced mitochondrial defects
T2D mouse provides an optimal experimental and apoptosis in vitro in dorsal root ganglia and
model, because these animals become hypergly- preserves nerve function in prediabetic HFD
cemic and dyslipidemic and develop PN [198, mice [167]. Oleate is thought to mediate these
199]. Treating db/db mice with pioglitazone, a neuroprotective effects through lipid droplet for-
peroxisome proliferator-activated receptor mation [167]. Polyunsaturated fatty acid diet
gamma (PPARγ) agonist, preserves nerve func- supplementation exerts similar protective effects
tion, lowers lipid levels, and alters expression of on PN in prediabetes and T2D rat models [203,
genes related to oxidative stress, fatty acid 204].
β-oxidation, and the tricarboxylic acid cycle We have also investigated the influence of
[188, 200, 201]. However, fluxomics experiments HFD in saphenous nerve in vivo in prediabetic
in sciatic nerves from db/db T2D mice offer the mice [29]. HFD affected the number, speed, and
best comparison of different energy substrates size of mitochondria trafficking in the antero-
[175, 202]. Administering db/db mice radiola- grade and retrograde directions. It also depolar-
beled [U-13C6] glucose revealed no change in gly- ized axonal mitochondria and impaired their
colysis, but a significant decrease in tricarboxylic ability to dissipate mitochondrial membrane
acid cycle intermediates in neuropathic sciatic potential in response to physiological demand.
nerve, underscoring mitochondrial dysfunction. Moreover, HFD lowered sensory axonal calcium
Using [13C16] palmitate, neuropathic sciatic levels, lengthened mitochondria, and increased
nerves had increased β-oxidation and acylcarni- PGC1α expression, overall suggesting that mito-
tine accumulation, followed by a drop in tricar- chondrial dysfunction and an imbalance of axo-
boxylic acid cycle substrates. Thus, excess nal energy underlies PN in prediabetes.
glucose and lipids alter mitochondrial bioener- Schwann cell metabolism is also susceptible to
getics in T2D nerves in distinct and complimen- hyperglycemic and dyslipidemic conditions.
tary ways. Future fluxomic studies in the Hyperglycemia induces Schwann cell oxidative
diet-induced HFD prediabetic model would shed stress, mitochondrial dysfunction, and apoptosis
more insight on nutrient flux and mitochondrial in vitro [205–208] and in vivo in diabetes animal
bioenergetics specifically in the dyslipidemia set- models of PN [209]. Dyslipidemia also elicits
ting, an avenue warranting investigation. Schwann cell injury; treatment with the saturated
Cell culture models have also proven useful fatty acid palmitate in vitro lowers Schwann cell
for increasing our understanding of mitochon- endoplasmic reticulum calcium stores, followed
drial bioenergetics and dynamics. Simulating by mitochondrial membrane depolarization, and
dyslipidemic conditions in vitro through long-­ an increase in oxidative stress [87]. Pretreatment
chain saturated fatty acid palmitate treatment with elevated glucose levels potentiates the detri-
depolarizes axonal mitochondria, reduces intra- mental effect of palmitate; thus, similar to the sce-
cellular ATP levels, induces oxidative stress, and nario in neurons, hyperglycemia and dyslipidemia
raises apoptosis of dorsal root ganglia neurons may synergize to induce Schwann cell damage.
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 297

In summary, given the high energy demands T1D and T2D participants [211]; however, one
of the PNS, neurons rely heavily on mitochon- limitation was that the study did not stratify par-
dria. Therefore, the susceptibility of mitochon- ticipants based on their diabetes status, i.e., T1D
dria to hyperglycemia and dyslipidemia on from T2D, and did not assess intraepidermal
bioenergetics, substrate utilization, biogenesis, nerve fiber density. The results from these four
trafficking, and reactive oxygen species produc- trials are promising, but call for confirmatory
tion ultimately lead to PN. studies on larger cohorts to validate the role of
exercise as a disease-modifying PN therapy.
Several additional studies have also investigated
4 Disease-Modifying the benefit of exercise on steadiness and gait, and
Therapies for PN Associated quality of life [reviewed in [210]]. Moving for-
with Dyslipidemia ward, research comparing the efficacy of aerobic
versus high-intensity interval training will be
Our increased understanding of the lipid-centric critical to fully elucidate the optimal exercise
molecular mechanisms underlying PN and the regimen for PN improvement.
emerging role of obesity as a causal factor advo-
cate lifestyle and dietary interventions as PN
treatments [131, 210]. Although large, random- 4.2 Dietary Interventions
ized controlled clinical trials are lacking, the
emergent evidence is that lifestyle interventions Besides exercise, preclinical studies strongly
may exert beneficial effects, and exercise is now support a role for dietary interventions for revers-
recommended for persons with T2D and PN by ing PN [26, 28, 167, 203, 204, 215]. Switching
the American Diabetes Association [30]. Here, prediabetic or T2D mice from a saturated to a
we will review the current level of evidence for monounsaturated fatty acid-rich diet reverses
lifestyle interventions as well as lipid-centric metrics of both small and large fiber PN, aligning
pharmacological approaches for PN. with the differences these fatty acids have on
mitochondrial bioenergetics [28, 167]. Although
studies on PN in humans are lacking, diets rich in
4.1 Exercise monounsaturated or polyunsaturated fatty acid
are beneficial to metabolic health and T2D and
The potential of exercise as a therapeutic strategy cardiovascular disease prevention [34, 216–218].
for persons with PN has been evaluated in three One small open-label study of omega-3 supple-
non-randomized and one small randomized trials mentation in T1D participants with PN improved
[211–214]. Intraepidermal nerve fiber density corneal nerve fiber length by corneal confocal
increased in patients with impaired glucose toler- microscopy [219]. However, corneal nerve fiber
ance and PN after an individualized 12-month length is not a validated PN metric; indeed, nei-
exercise intervention program along with diet ther nerve conduction nor cooling or vibration
[214]. More recent trials showed similar improve- threshold improved upon omega-3
ments in cutaneous reinnervation rates and nerve supplementation.
fiber branching in PN following aerobic exercise Preclinical studies further demonstrate a link
intervention in prediabetes and diabetes partici- between fatty acid dietary source and nerve func-
pants [212, 213]. Interestingly, these interven- tion [28, 167], a finding that was confirmed in the
tions were not associated with significant weight Netherlands Epidemiology of Obesity (NEO)
loss and used intraepidermal nerve fiber density study, which found a positive correlation between
as a primary outcome measure. Balducci et  al. animal-based but not dairy- or plant-based satu-
showed improved nerve conduction velocity and rated and unsaturated fatty acids with markers of
vibration perception after a 4-year aerobic exer- insulin resistance [220]. Consistent with these
cise intervention in a small randomized trial of findings, patients with T2D and painful PN
298 S. A. Eid et al.

placed on a low-fat, plant-based diet experienced sity interval training, alone or combined, in a ran-
lower pain symptoms after 20 weeks of interven- domized trial on PN in obese participants
tion [221]. However, firm conclusions could not (NCT03617185) [2]. This study will utilize the
be drawn because these participants also lost gold standard intraepidermal nerve fiber density
weight, which likely contributed to improvement as the primary outcome and constitutes a critical
in nerve function. Although data on the effects of step to determine if either intervention, bariatric
dietary-only interventions in PN are sparse, we surgery or high-intensity exercise, will generate
posit that a plant-based diet enriched in unsatu- superior PN outcomes.
rated fatty acids with or without exercise is essen-
tial for peripheral nerve health and function, in
line with the recommendations of the American 4.4 Lipid-Lowering Agents
Diabetes Association [30].
In terms of pharmacological interventions, it
remains unclear whether single- or multi-­factorial
4.3 Weight Loss approaches are needed to achieve sufficient thera-
peutic efficacy. Incorporating lipid-lowering
Interventions designed to evaluate the effect of agents may be effective for treating PN. Fenofibrate,
medical or surgical weight loss on PN are lim- a PPARα agonist, which reduces plasma triglycer-
ited. Recently, the Look AHEAD (Action for ides and modestly increases HDL-c [128], has
Health in Diabetes) trial, which aimed to achieve shown promise for treating diabetic microvascular
and maintain long-term weight loss through complications [225, 226], including PN [227]. The
intensive lifestyle intervention program, showed Fenofibrate Intervention and Event Lowering in
PN improvement in overweight and obese Diabetes (FIELD) study showed that fenofibrate
patients with T2D according to the Michigan treatment significantly reduced the risk of lower-
Neuropathy Screening Instrument questionnaire limb amputations in T2D patients [227]. Additional
and light touch sensation [222]. The benefits, mechanistic studies in db/db mice demonstrated
however, mostly did not persist 1–2.3 years after that these neuroprotective properties may be medi-
study termination. Another long-term r­ andomized ated by the AMP activated protein kinase (AMPK)-
Diabetes Prevention Program Outcome Study PGC1α pathway [228], an effect that needs to be
(DPPOS) compared lifestyle intervention to met- validated in other T2D mouse models and in
formin or placebo [223]. At a mean 15-year fol- human T2D PN.
low-up, there were no differences among groups On the other hand, the efficacy of statin ther-
for composite PN, nephropathy, and retinopathy apy, aimed at reducing plasma LDL-c, remains
outcomes, although there was a trending decrease controversial. While Rajamani et al. showed that
in PN in the lifestyle group. When stratified by statin use reduces the risk of developing PN and
sex, lifestyle intervention significantly lowered foot ulcers in T2D patients [229], increasingly,
composite microvascular outcomes in females. data reveal a positive association between pro-
Additionally, an ongoing clinical trial by our longed statin treatment duration and elevated PN
group will shed light on the efficacy of medical risk [230, 231]. Additionally, a recent review sug-
weight loss on PN progression in obese and pre- gested a potential link between statin therapy and
diabetic participants (NCT02689661) [4]. the development of neuropathic pain [232]. Our
Regarding surgical weight loss, a small pro- large population-based study of 259,625 T2D
spective study of T2D participants found that participants found that statin therapy neither
Roux-en-Y gastric bypass surgery improved PN worsened nor improved PN, although it could not
at 6-month follow-up, according to the discount a small acute risk of harm [233]. Thus,
Neuropathy Symptom Score and the Neuropathy more mechanistic studies are needed to deter-
Deficit Score [224]. We are also examining the mine if statin use is a meaningful therapeutic PN
effects of bariatric surgery as well as high inten- strategy.
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 299

Lastly, it is important to emphasize that these intersection between glucose- and lipid-centric
drugs are not tissue-specific and may have dif- mechanisms in PN and axo-glial metabolic com-
ferential effects on complication-prone tissues, in munication and the non-cell autonomous nature
addition to pleiotropic actions independent of of PN. Adoption of omics and multi-omics tech-
lipid-lowering [234, 235]. Thus, besides drug niques followed by target validation will advance
refinement, identifying cellular and subcellular our understanding of pathophysiology and iden-
targets of lipotoxicity may be the key to attain tify actionable targets for developing disease-­
full efficacy against nerve damage. Of interest, modifying therapy for PN secondary to metabolic
we recently applied network analysis of per- dysfunction. Additionally, the contribution of the
turbed and interacting pathways to identify metabolic syndrome, obesity, and dyslipidemia
PN-induced rewiring between pathogenic path- to PN in the T1D setting will need to be addressed
ways, which offers alternative pharmacological as obesity rises in this population as well.
strategies for PN treatment [236]. Cross-species Presently, diet, weight loss, and exercise are
comparison of human versus mouse transcrip- recommended as the first-line PN treatment to
tomes (detailed above) is another critical reverse metabolic syndrome components and
approach for offering mechanism-based targets improve glucose handling and lipid profiles,
conserved across mouse and human and improv- anticipating this will prevent, slow, or reverse PN
ing therapeutic interventions [165]. [30]. Preliminary clinical evidence indicates
these may be promising avenues. Indeed, life-
style interventions have long been regarded as
5 Conclusions beneficial for T2D patients, as one review stated
even as long as 20 years ago, “The question is no
The past decade has seen PN research move past longer whether diet and exercise can benefit the
the purely glucose-centric view to also encom- individual with type 2 diabetes” but rather to
pass lipid-centric concepts. This parallels fre- what extent and on the “…appropriate feasibility
quent comorbid obesity and dyslipidemia present and acceptability of an intervention program.”
in prediabetes and T2D patients, and the complex We await the findings from randomized, con-
intersection of glucose and lipid metabolism at trolled clinical trials, which will evaluate the effi-
the molecular level. Large clinical studies have cacy of diet, exercise, and surgical weight loss on
outlined that these additional risk factors for PN, PN to identify the optimal regimen.
obesity, dyslipidemia, and the metabolic syn-
drome, can also occur independent of hypergly- Acknowledgments The authors thank Bhumsoo Kim,
cemia. The clinical findings also suggest that PhD, for revising content and for critical input.
glucose control alone may not suffice to prevent
PN onset or progression in T2D patients, and that
the metabolic syndrome is a critical driver of PN References
pathogenesis. Preclinical studies are uncovering
1. Callaghan BC, et  al. Enhanced glucose control
the pathomechanisms of dyslipidemia on nerves for preventing and treating diabetic neuropathy.
through lipogenesis, mitochondrial bioenergetics Cochrane Database Syst Rev. 2012;6:CD007543.
and transport, as well as oxidative stress and 2. Callaghan BC, et  al. Central obesity is associated
inflammation. A clarification of the mechanisms with neuropathy in the severely obese. Mayo Clin
Proc. 2020;95(7):1342–53.
in the appropriate models are needed, since many 3. Callaghan BC, et  al. Metabolic syndrome compo-
previous studies utilized T1D or T2D animals nents are associated with symptomatic polyneuropa-
possessing genetic mutations, e.g., leptin signal- thy independent of glycemic status. Diabetes Care.
ing deficient db/db [22], which do not fully model 2016;39(5):801–7.
4. Callaghan BC, et  al. Association between meta-
the human condition [25]. bolic syndrome components and polyneuropa-
Additionally, emergent research avenues thy in an obese population. JAMA Neurol.
require deeper understanding, including the 2016;73(12):1468–76.
300 S. A. Eid et al.

5. Jaiswal M, et al. Prevalence of and risk factors for 20. Ismail-Beigi F, et al. Effect of intensive treatment of
diabetic peripheral neuropathy in youth with type 1 hyperglycaemia on microvascular outcomes in type
and type 2 diabetes: SEARCH for Diabetes in Youth 2 diabetes: an analysis of the ACCORD randomised
Study. Diabetes Care. 2017;40(9):1226–32. trial. Lancet. 2010;376(9739):419–30.
6. Jaiswal M, et  al. Burden of diabetic peripheral 21. Callaghan BC, et  al. The prevalence and determi-
neuropathy in pima indians with type 2 diabetes. nants of cognitive deficits and traditional diabetic
Diabetes Care. 2016;39(4):e63–4. complications in the severely obese. Diabetes Care.
7. Callaghan BC, et  al. Triglycerides and amputa- 2020;43(3):683–90.
tion risk in patients with diabetes: ten-year fol- 22. O'Brien PD, Sakowski SA, Feldman EL.  Mouse
low-­up in the DISTANCE study. Diabetes Care. models of diabetic neuropathy. ILAR J.
2011;34(3):635–40. 2014;54(3):259–72.
8. Smith AG, Singleton JR.  Obesity and hyperlipid- 23. Elzinga SE, et  al. Sex differences in insulin resis-
emia are risk factors for early diabetic neuropathy. J tance but not peripheral neuropathy in a diet-­
Diabetes Complicat. 2013;27(5):436–42. induced prediabetes mouse model. Dis Model Mech.
9. Callaghan BC, et  al. Diabetes and obesity are the 2021;14(4):48909.
main metabolic drivers of peripheral neuropathy. 24. Medrikova D, et  al. Sex differences during the
Ann Clin Transl Neurol. 2018;5(4):397–405. course of diet-induced obesity in mice: adipose tis-
10. Lu B, et  al. Determination of peripheral neuropa- sue expandability and glycemic control. Int J Obes.
thy prevalence and associated factors in Chinese 2012;36(2):262–72.
subjects with diabetes and pre-diabetes - ShangHai 25. Farr OM, Gavrieli A, Mantzoros CS.  Leptin appli-
Diabetic neuRopathy Epidemiology and Molecular cations in 2015: what have we learned about leptin
Genetics Study (SH-DREAMS). PLoS One. and obesity? Curr Opin Endocrinol Diabetes Obes.
2013;8(4):e61053. 2015;22(5):353–9.
11. Han L, et  al. Peripheral neuropathy is associated 26. Hinder LM, et al. Dietary reversal of neuropathy in
with insulin resistance independent of metabolic a murine model of prediabetes and metabolic syn-
syndrome. Diabetol Metab Syndr. 2015;7:14. drome. Dis Model Mech. 2017;10(6):717–25.
12. Andersen ST, et al. Risk factors for incident diabetic 27. O’Brien PD, et  al. Juvenile murine models of pre-
polyneuropathy in a cohort with screen-detected diabetes and type 2 diabetes develop neuropathy. Dis
type 2 diabetes followed for 13 years: ADDITION-­ Model Mech. 2018;11(12):37374.
Denmark. Diabetes Care. 2018;41(5):1068–75. 28. O'Brien PD, et al. Integrated lipidomic and transcrip-
13. Christensen DH, et  al. Metabolic factors, life- tomic analyses identify altered nerve triglycerides in
style habits, and possible polyneuropathy in early mouse models of prediabetes and type 2 diabetes.
type 2 diabetes: a nationwide study of 5,249 Dis Model Mech. 2020;13(2):42101.
patients in the danish centre for strategic research 29. Sajic M, et al. High dietary fat consumption impairs
in type 2 diabetes (DD2) cohort. Diabetes Care. axonal mitochondrial function in  vivo. J Neurosci.
2020;43(6):1266–75. 2021;41(19):4321–34.
14. Schlesinger S, et al. General and abdominal obesity 30. Pop-Busui R, et al. Diabetic neuropathy: a position
and incident distal sensorimotor polyneuropathy: statement by the American Diabetes Association.
insights into inflammatory biomarkers as potential Diabetes Care. 2017;40(1):136–54.
mediators in the KORA F4/FF4 cohort. Diabetes 31. Tuomilehto J, et  al. Prevention of type 2 diabetes
Care. 2019;42(2):240–7. mellitus by changes in lifestyle among subjects
15. Ziegler D, et  al. Prevalence of polyneuropathy in with impaired glucose tolerance. N Engl J Med.
pre-diabetes and diabetes is associated with abdomi- 2001;344(18):1343–50.
nal obesity and macroangiopathy: the MONICA/ 32. Aucott L, et al. Effects of lifestyle interventions and
KORA Augsburg Surveys S2 and S3. Diabetes Care. long-term weight loss on lipid outcomes - a system-
2008;31(3):464–9. atic review. Obes Rev. 2011;12(5):e412–25.
16. Hanewinckel R, et  al. High body mass and kidney 33. Tjønna AE, et  al. Aerobic interval training versus
dysfunction relate to worse nerve function, even in continuous moderate exercise as a treatment for
adults without neuropathy. J Peripher Nerv Syst. the metabolic syndrome: a pilot study. Circulation.
2017;22(2):112–20. 2008;118(4):346–54.
17. Grisold A, Callaghan BC, Feldman EL.  Mediators 34. Salas-Salvado J, et al. Reduction in the incidence of
of diabetic neuropathy: is hyperglycemia the only type 2 diabetes with the Mediterranean diet: results
culprit? Curr Opin Endocrinol Diabetes Obes. of the PREDIMED-Reus nutrition intervention ran-
2017;24(2):103–11. domized trial. Diabetes Care. 2011;34(1):14–9.
18. Stino AM, Smith AG. Peripheral neuropathy in pre- 35. Sasaki Y.  Metabolic aspects of neuronal degenera-
diabetes and the metabolic syndrome. J Diabetes tion: from a NAD+ point of view. Neurosci Res.
Investig. 2017;8(5):646–55. 2019;139:9–20.
19. Duckworth W, et  al. Glucose control and vascular 36. Feldman EL, et al. New horizons in diabetic neurop-
complications in veterans with type 2 diabetes. N athy: mechanisms, bioenergetics, and pain. Neuron.
Engl J Med. 2009;360(2):129–39. 2017;93(6):1296–313.
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 301

37. Wang JT, Medress ZA, Barres BA.  Axon degen- 57. Kastaniotis AJ, et  al. Mitochondrial fatty acid
eration: molecular mechanisms of a self-destruction synthesis, fatty acids and mitochondrial physiol-
pathway. J Cell Biol. 2012;196(1):7–18. ogy. Biochim Biophys Acta Mol Cell Biol Lipids.
38. Rossi MJ, Pekkurnaz G.  Powerhouse of the mind: 2017;1862(1):39–48.
mitochondrial plasticity at the synapse. Curr Opin 58. Kidd GJ, Ohno N, Trapp BD. Biology of Schwann
Neurobiol. 2019;57:149–55. cells. Handb Clin Neurol. 2013;115:55–79.
39. Zhu XH, et  al. Quantitative imaging of energy 59. Schulingkamp RJ, et al. Insulin receptors and insulin
expenditure in human brain. NeuroImage. action in the brain: review and clinical implications.
2012;60(4):2107–17. Neurosci Biobehav Rev. 2000;24(8):855–72.
40. Mitchell RW, et  al. Fatty acid transport protein 60. Jha MK, Morrison BM. Lactate transporters mediate
expression in human brain and potential role in fatty glia-neuron metabolic crosstalk in homeostasis and
acid transport across human brain microvessel endo- disease. Front Cell Neurosci. 2020;14:589582.
thelial cells. J Neurochem. 2011;117(4):735–46. 61. Magnani P, et al. Glucose transporters in rat periph-
41. Poitelon Y, Kopec AM, Belin S. Myelin fat facts: an eral nerve: paranodal expression of GLUT1 and
overview of lipids and fatty acid metabolism. Cell. GLUT3. Metabolism. 1996;45(12):1466–73.
2020;9(4):812. 62. Magnani P, et  al. Regulation of glucose transport
42. Simpson IA, et  al. The facilitative glucose trans- in cultured Schwann cells. J Peripher Nerv Syst.
porter GLUT3: 20 years of distinction. Am J Physiol 1998;3(1):28–36.
Endocrinol Metab. 2008;295(2):E242–53. 63. Jha MK, Morrison BM. Glia-neuron energy metabo-
43. Bender T, Martinou JC. The mitochondrial pyruvate lism in health and diseases: New insights into the
carrier in health and disease: to carry or not to carry? role of nervous system metabolic transporters. Exp
Biochim Biophys Acta. 2016;1863(10):2436–42. Neurol. 2018;309:23–31.
44. Krebs HA, Johnson WA. Metabolism of ketonic acids 64. Brown AM, et al. Schwann cell glycogen selectively
in animal tissues. Biochem J. 1937;31(4):645–60. supports myelinated axon function. Ann Neurol.
45. Chaban Y, Boekema EJ, Dudkina NV.  Structures 2012;72(3):406–18.
of mitochondrial oxidative phosphorylation super- 65. Véga C, et  al. Uptake of locally applied deoxyglu-
complexes and mechanisms for their stabilisation. cose, glucose and lactate by axons and Schwann
Biochim Biophys Acta. 2014;1837(4):418–26. cells of rat vagus nerve. J Physiol. 2003;546(Pt
46. Cecchini G.  Function and structure of complex 2):551–64.
II of the respiratory chain. Annu Rev Biochem. 66. Bouçanova F, Chrast R.  Metabolic interaction
2003;72:77–109. between schwann cells and axons under physiologi-
47. Hirst J. Energy transduction by respiratory complex cal and disease conditions. Front Cell Neurosci.
I–an evaluation of current knowledge. Biochem Soc 2020;14:148.
Trans. 2005;33(Pt 3):525–9. 67. Gonçalves NP, et al. Schwann cell interactions with
48. Crofts AR.  The cytochrome bc1 complex: func- axons and microvessels in diabetic neuropathy. Nat
tion in the context of structure. Annu Rev Physiol. Rev Neurol. 2017;13(3):135–47.
2004;66:689–733. 68. Domenech-Estevez E, et  al. Distribution of mono-
49. Tsukihara T, et  al. The whole structure of the carboxylate transporters in the peripheral nervous
13-subunit oxidized cytochrome c oxidase at 2.8 system suggests putative roles in lactate shuttling
A. Science. 1996;272(5265):1136–44. and myelination. J Neurosci. 2015;35(10):4151–6.
50. Boyer PD. The ATP synthase–a splendid molecular 69. Babetto E, Wong KM, Beirowski B.  A glycolytic
machine. Annu Rev Biochem. 1997;66:717–49. shift in Schwann cells supports injured axons. Nat
51. Rangaraju V, Calloway N, Ryan TA. Activity-driven Neurosci. 2020;23(10):1215–28.
local ATP synthesis is required for synaptic function. 70. Morrison BM, et al. Deficiency in monocarboxylate
Cell. 2014;156(4):825–35. transporter 1 (MCT1) in mice delays regeneration of
52. Sobieski C, Fitzpatrick MJ, Mennerick peripheral nerves following sciatic nerve crush. Exp
SJ.  Differential presynaptic ATP supply for basal Neurol. 2015;263:325–38.
and high-demand transmission. J Neurosci. 71. Bouçanova F, et  al. Disrupted function of lactate
2017;37(7):1888–99. transporter MCT1, but not MCT4, in Schwann cells
53. Grote CW, Ryals JM, Wright DE. In vivo peripheral affects the maintenance of motor end-plate innerva-
nervous system insulin signaling. J Peripher Nerv tion. Glia. 2021;69(1):124–36.
Syst. 2013;18(3):209–19. 72. Jha MK, et  al. Reducing monocarboxylate trans-
54. Grote CW, et  al. Peripheral nervous system insu- porter MCT1 worsens experimental diabetic periph-
lin resistance in ob/ob mice. Acta Neuropathol eral neuropathy. Exp Neurol. 2020;333:113415.
Commun. 2013;1:15. 73. Jha MK, et  al. Monocarboxylate transporter 1  in
55. Grote CW, Wright DE. A role for insulin in diabetic Schwann cells contributes to maintenance of
neuropathy. Front Neurosci. 2016;10:581. sensory nerve myelination during aging. Glia.
56. Houten SM, Wanders RJ. A general introduction to 2020;68(1):161–77.
the biochemistry of mitochondrial fatty acid beta-­ 74. Rumora AE, et  al. Disorders of mitochondrial
oxidation. J Inherit Metab Dis. 2010;33(5):469–77. dynamics in peripheral neuropathy: Clues from
302 S. A. Eid et al.

hereditary neuropathy and diabetes. Int Rev 93. Trecarichi A, Flatters SJL.  Mitochondrial dys-
Neurobiol. 2019;145:127–76. function in the pathogenesis of chemotherapy-­
75. Courchet J, et  al. Terminal axon branching is induced peripheral neuropathy. Int Rev Neurobiol.
regulated by the LKB1-NUAK1 kinase path- 2019;145:83–126.
way via presynaptic mitochondrial capture. Cell. 94. Angelova PR, Abramov AY.  Role of mitochondrial
2013;153(7):1510–25. ROS in the brain: from physiology to neurodegen-
76. Spillane M, et  al. Mitochondria coordinate sites of eration. FEBS Lett. 2018;592(5):692–702.
axon branching through localized intra-axonal pro- 95. Oyewole AO, Birch-Machin MA.  Mitochondria-­
tein synthesis. Cell Rep. 2013;5(6):1564–75. targeted antioxidants. FASEB J.
77. Sun T, et al. Motile axonal mitochondria contribute 2015;29(12):4766–71.
to the variability of presynaptic strength. Cell Rep. 96. Snezhkina AV, et  al. ROS generation and antioxi-
2013;4(3):413–9. dant defense systems in normal and malignant cells.
78. Sheng Z-H. The interplay of axonal energy homeo- Oxidative Med Cell Longev. 2019;2019:6175804.
stasis and mitochondrial trafficking and anchoring. 97. Akude E, et  al. Diminished superoxide genera-
Trends Cell Biol. 2017;27(6):403–16. tion is associated with respiratory chain dysfunc-
79. Amiri M, Hollenbeck PJ. Mitochondrial biogenesis tion and changes in the mitochondrial proteome
in the axons of vertebrate peripheral neurons. Dev of sensory neurons from diabetic rats. Diabetes.
Neurobiol. 2008;68(11):1348–61. 2011;60(1):288–97.
80. Schwarz TL.  Mitochondrial trafficking in neurons. 98. Nishikawa T, et al. Normalizing mitochondrial super-
Cold Spring Harb Perspect Biol. 2013;5(6):11304. oxide production blocks three pathways of hypergly-
81. Lentz SI, et  al. Mitochondrial DNA (mtDNA) bio- caemic damage. Nature. 2000;404(6779):787–90.
genesis: visualization and duel incorporation of 99. Vincent AM, et  al. Short-term hyperglycemia pro-
BrdU and EdU into newly synthesized mtDNA duces oxidative damage and apoptosis in neurons.
in vitro. J Histochem Cytochem. 2010;58(2):207–18. FASEB J. 2005;19(6):638–40.
82. Yu SB, Pekkurnaz G.  Mechanisms orchestrating 100. Tretter L, Adam-Vizi V.  Alpha-ketoglutarate dehy-
mitochondrial dynamics for energy homeostasis. J drogenase: a target and generator of oxidative stress.
Mol Biol. 2018;430(21):3922–41. Philos Trans R Soc B. 2005;360(1464):2335–45.
83. Rumora AE, et  al. Chain length of saturated fatty 101. Gardner P, Fridovich I.  Inactivation-reactivation
acids regulates mitochondrial trafficking and function of aconitase in Escherichia coli. A sensitive
in sensory neurons. J Lipid Res. 2019;60(1):58–70. measure of superoxide radical. J Biol Chem.
84. Rumora AE, et al. Dyslipidemia impairs mitochon- 1992;267(13):8757–63.
drial trafficking and function in sensory neurons. 102. Vincent AM, et  al. Sensory neurons and Schwann
FASEB J. 2018;32(1):195–207. cells respond to oxidative stress by increasing
85. Hinder LM, et  al. Long-chain acyl coenzyme A antioxidant defense mechanisms. Antioxid Redox
synthetase 1 overexpression in primary cultured Signal. 2009;11(3):425–38.
Schwann cells prevents long chain fatty acid-­ 103. Delaney CL, et  al. Insulin-like growth factor-I and
induced oxidative stress and mitochondrial dysfunc- over-expression of Bcl-xL prevent glucose-mediated
tion. Antioxid Redox Signal. 2014;21(4):588–600. apoptosis in Schwann cells. J Neuropathol Exp
86. Viader A, et  al. Aberrant Schwann cell lipid Neurol. 2001;60(2):147–60.
metabolism linked to mitochondrial deficits leads 104. Thurston JH, et  al. Effects of acute, subacute, and
to axon degeneration and neuropathy. Neuron. chronic diabetes on carbohydrate and energy metab-
2013;77(5):886–98. olism in rat sciatic nerve: relation to mechanisms of
87. Padilla A, et  al. Hyperglycemia magnifies peripheral neuropathy. Diabetes. 1995;44(2):190–5.
Schwann cell dysfunction and cell death trig- 105. Kishi Y, et  al. Alpha-lipoic acid: effect on glucose
gered by PA-induced lipotoxicity. Brain Res. uptake, sorbitol pathway, and energy metabolism
2011;1370:64–79. in experimental diabetic neuropathy. Diabetes.
88. Nicholls DG, Budd SL. Mitochondria and neuronal 1999;48(10):2045–51.
survival. Physiol Rev. 2000;80(1):315–60. 106. Obrosova IG, et  al. Aldose reductase inhibi-
89. Fernyhough P.  Mitochondrial dysfunction in dia- tion counteracts oxidative-nitrosative stress and
betic neuropathy: a series of unfortunate metabolic poly (ADP-ribose) polymerase activation in tis-
events. Curr Diab Rep. 2015;15(11):89. sue sites for diabetes complications. Diabetes.
90. Chowdhury SK, Smith DR, Fernyhough P. The role 2005;54(1):234–42.
of aberrant mitochondrial bioenergetics in diabetic 107. Yagihashi S.  Glucotoxic mechanisms and related
neuropathy. Neurobiol Dis. 2013;51:56–65. therapeutic approaches. Int Rev Neurobiol.
91. Vincent AM, et  al. Mitochondrial biogenesis and 2016;127:121–49.
fission in axons in cell culture and animal mod- 108. Dewanjee S, et al. Molecular mechanism of diabetic
els of diabetic neuropathy. Acta Neuropathol. neuropathy and its pharmacotherapeutic targets. Eur
2010;120(4):477–89. J Pharmacol. 2018;833:472–523.
92. Colvin LA. Chemotherapy-induced peripheral neu- 109. Greene DA, Lattimer S.  Impaired rat sciatic nerve
ropathy: where are we now? Pain. 2019;160(Suppl sodium-potassium adenosine triphosphatase in
1):1–10. acute streptozocin diabetes and its correction by
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 303

dietary myo-inositol supplementation. J Clin Invest. of randomized clinical trials. Diabetes Metab J.
1983;72(3):1058–63. 2013;37(5):375.
110. Greene DA, Lattimer SA, Sima AA. Sorbitol, phos- 127. Callaghan BC, Hur J, Feldman EL.  Diabetic neu-
phoinositides, and sodium-potassium-ATPase in the ropathy: one disease or two? Curr Opin Neurol.
pathogenesis of diabetic complications. N Engl J 2012;25(5):536–41.
Med. 1987;316(10):599–606. 128. Vincent AM, et al. Hyperlipidemia: a new therapeu-
111. Greene DA, et  al. Nerve Na+-K+-ATPase, tic target for diabetic neuropathy. J Peripher Nerv
conduction, and myo-inositol in the insulin-­ Syst. 2009;14(4):257–67.
deficient BB rat. Am J Physiol Endocrinol Metab. 129. Wiggin TD, et  al. Elevated triglycerides correlate
1984;247(4):E534–9. with progression of diabetic neuropathy. Diabetes.
112. Oates PJ.  Aldose reductase, still a compelling tar- 2009;58(7):1634–40.
get for diabetic neuropathy. Curr Drug Targets. 130. Eid S, et  al. New insights into the mechanisms of
2008;9(1):14–36. diabetic complications: role of lipids and lipid
113. Stevens MJ, et  al. The linked roles of nitric oxide, metabolism. Diabetologia. 2019;62(9):1539–49.
aldose reductase and (Na+, K+)-ATPase in the slow- 131. Savelieff MG, Callaghan BC, Feldman EL.  The
ing of nerve conduction in the streptozotocin dia- emerging role of dyslipidemia in diabetic microvas-
betic rat. J Clin Invest. 1994;94(2):853–9. cular complications. Curr Opin Endocrinol Diabetes
114. Kern TS, Engerman RL.  Immunohistochemical Obes. 2020;27(2):115–23.
distribution of aldose reductase. Histochem J. 132. Franssen R, et  al. Obesity and dyslipidemia. Med
1982;14(3):507–15. Clin North Am. 2011;95(5):893–902.
115. Cameron N, Cotter M.  Potential therapeutic 133. Corbin KD, et al. Obesity in type 1 diabetes: patho-
approaches to the treatment or prevention of diabetic physiology, clinical impact, and mechanisms.
neuropathy: evidence from experimental studies. Endocr Rev. 2018;39(5):629–63.
Diabet Med. 1993;10(7):593–605. 134. Mottalib A, et  al. Weight management in patients
116. Ho EC, et  al. Aldose reductase–deficient mice are with type 1 diabetes and obesity. Curr Diab Rep.
protected from delayed motor nerve conduction 2017;17(10):92.
velocity, increased c-Jun NH2-terminal kinase acti- 135. O'Brien PD, et  al. Neurological consequences of
vation, depletion of reduced glutathione, increased obesity. Lancet Neurol. 2017;16(6):465–77.
superoxide accumulation, and DNA damage. 136. Ebbert JO, Jensen MD. Fat depots, free fatty acids,
Diabetes. 2006;55(7):1946–53. and dyslipidemia. Nutrients. 2013;5(2):498–508.
117. Yagihashi S, et  al. Neuropathy in diabetic mice 137. Tchoukalova YD, et  al. Regional differences
overexpressing human aldose reductase and in cellular mechanisms of adipose tissue gain
effects of aldose reductase inhibitor. Brain. with overfeeding. Proc Natl Acad Sci U S A.
2001;124(12):2448–58. 2010;107(42):18226–31.
118. Tsai SC, Burnakis TG. Aldose reductase inhibitors: 138. Acosta JR, et al. Increased fat cell size: a major phe-
an update. Ann Pharmacother. 1993;27(6):751–4. notype of subcutaneous white adipose tissue in non-­
119. Chalk C, Benstead TJ, Moore F.  Aldose reductase obese individuals with type 2 diabetes. Diabetologia.
inhibitors for the treatment of diabetic polyneuropa- 2016;59(3):560–70.
thy. Cochrane Database Syst Rev. 2007;4:CD004572. 139. Wensveen FM, et  al. The “Big Bang” in obese
120. Misur I, et  al. Advanced glycation endproducts in fat: events initiating obesity-induced adi-
peripheral nerve in type 2 diabetes with neuropathy. pose tissue inflammation. Eur J Immunol.
Acta Diabetol. 2004;41(4):158–66. 2015;45(9):2446–56.
121. Popova EA, Mironova RS, Odjakova MK.  Non-­ 140. Fontana L, et  al. Visceral fat adipokine secretion
enzymatic glycosylation and deglycating enzymes. is associated with systemic inflammation in obese
Biotechnol Biotechnol Equip. 2010;24(3):1928–35. humans. Diabetes. 2007;56(4):1010–3.
122. Lukic IK, et al. The RAGE pathway: activation and 141. Vincent AM, et  al. Dyslipidemia-induced neuropa-
perpetuation in the pathogenesis of diabetic neurop- thy in mice: the role of oxLDL/LOX-1. Diabetes.
athy. Ann N Y Acad Sci. 2008;1126(1):76–80. 2009;58(10):2376–85.
123. Du X-L, et al. Hyperglycemia-induced mitochondrial 142. Ouchi N, et  al. Adipokines in inflamma-
superoxide overproduction activates the hexosamine tion and metabolic disease. Nat Rev Immunol.
pathway and induces plasminogen activator inhibi- 2011;11(2):85–97.
tor-­1 expression by increasing Sp1 glycosylation. 143. Bartness TJ, et al. Sensory and sympathetic nervous
Proc Natl Acad Sci U S A. 2000;97(22):12222–6. system control of white adipose tissue lipolysis. Mol
124. Eichberg J.  Protein kinase C changes in diabe- Cell Endocrinol. 2010;318(1-2):34–43.
tes: is the concept relevant to neuropathy? Int Rev 144. Unamuno X, et  al. Adipokine dysregulation and
Neurobiol. 2002;50:61–82. adipose tissue inflammation in human obesity. Eur J
125. Geraldes P, King GL. Activation of protein kinase C Clin Investig. 2018;48(9):e12997.
isoforms and its impact on diabetic complications. 145. Schweiger M, et al. Adipose triglyceride lipase and
Circ Res. 2010;106(8):1319–31. hormone-sensitive lipase are the major enzymes
126. Bansal D, et  al. Ruboxistaurin for the treatment of in adipose tissue triacylglycerol catabolism. J Biol
diabetic peripheral neuropathy: a systematic review Chem. 2006;281(52):40236–41.
304 S. A. Eid et al.

146. Chen SC, Tseng CH. Dyslipidemia, kidney disease, 163. Hazra S, et  al. Liver X receptor modulates dia-
and cardiovascular disease in diabetic patients. Rev betic retinopathy outcome in a mouse model
Diabet Stud. 2013;10(2-3):88–100. of streptozotocin-induced diabetes. Diabetes.
147. Bardini G, Rotella CM, Giannini S.  Dyslipidemia 2012;61(12):3270–9.
and diabetes: reciprocal impact of impaired lipid 164. Gavini CK, et  al. Liver X receptors protect dorsal
metabolism and Beta-cell dysfunction on micro- root ganglia from obesity-induced endoplasmic
and macrovascular complications. Rev Diabet Stud. reticulum stress and mechanical allodynia. Cell Rep.
2012;9(2-3):82–93. 2018;25(2):271–277 e4.
148. Hotamisligil GS, Shargill NS, Spiegelman 165. McGregor BA, et al. Conserved transcriptional sig-
BM.  Adipose expression of tumor necrosis factor-­ natures in human and murine diabetic peripheral
alpha: direct role in obesity-linked insulin resistance. neuropathy. Sci Rep. 2018;8(1):17678.
Science. 1993;259(5091):87–91. 166. Eid SA, et  al. Targeting the NADPH oxidase-
149. Czech MP, et  al. Insulin signalling mecha- 4 and liver X receptor pathway preserves
­
nisms for triacylglycerol storage. Diabetologia. schwann cell integrity in diabetic mice. Diabetes.
2013;56(5):949–64. 2020;69(3):448–64.
150. Rhee EP, et al. Lipid profiling identifies a triacylg- 167. Rumora AE, et  al. The divergent roles of dietary
lycerol signature of insulin resistance and improves saturated and monounsaturated fatty acids on nerve
diabetes prediction in humans. J Clin Invest. function in murine models of obesity. J Neurosci.
2011;121(4):1402–11. 2019;39(19):3770–81.
151. Basu Ball W, Neff JK, Gohil VM. The role of non- 168. Wang H, Eckel RH.  Lipoprotein lipase: from
bilayer phospholipids in mitochondrial structure and gene to obesity. Am J Physiol Endocrinol Metab.
function. FEBS Lett. 2018;592(8):1273–90. 2009;297(2):E271–88.
152. Spector AA, Yorek MA.  Membrane lipid com- 169. Verheijen MH, et  al. Local regulation of fat
position and cellular function. J Lipid Res. metabolism in peripheral nerves. Genes Dev.
1985;26(9):1015–35. 2003;17(19):2450–64.
153. Djemli-Shipkolye A, et al. Na, K-atpase alterations 170. Pande M, et al. Transcriptional profiling of diabetic
in diabetic rats: relationship with lipid metabolism neuropathy in the BKS db/db mouse: a model of
and nerve physiological parameters. Cell Mol Biol. type 2 diabetes. Diabetes. 2011;60(7):1981–9.
2001;47(2):297–304. 171. Hur J, et  al. The identification of gene expres-
154. Montani L, et  al. De novo fatty acid synthe- sion profiles associated with progression of
sis by Schwann cells is essential for periph- human diabetic neuropathy. Brain. 2011;134(Pt
eral nervous system myelination. J Cell Biol. 11):3222–35.
2018;217(4):1353–68. 172. Supruniuk E, Miklosz A, Chabowski A. The impli-
155. Liang G, Cline GW, Macica CM. IGF-1 stimulates cation of PGC-1alpha on fatty acid transport across
de novo fatty acid biosynthesis by Schwann cells plasma and mitochondrial membranes in the insulin
during myelination. Glia. 2007;55(6):632–41. sensitive tissues. Front Physiol. 2017;8:923.
156. Camargo N, Smit AB, Verheijen MH.  SREBPs: 173. DeFronzo R.  Insulin resistance, lipotoxicity, type
SREBP function in glia-neuron interactions. FEBS 2 diabetes and atherosclerosis: the missing links.
J. 2009;276(3):628–36. The Claude Bernard Lecture 2009. Diabetologia.
157. Shimano H, Sato R. SREBP-regulated lipid metabo- 2010;53(7):1270–87.
lism: convergent physiology - divergent pathophysi- 174. D'Souza K, Nzirorera C, Kienesberger PC.  Lipid
ology. Nat Rev Endocrinol. 2017;13(12):710–30. metabolism and signaling in cardiac lipotoxicity.
158. Wang B, Tontonoz P.  Liver X receptors in lipid Biochim Biophys Acta. 2016;1861(10):1513–24.
signalling and membrane homeostasis. Nat Rev 175. Sas KM, et al. Shared and distinct lipid-lipid inter-
Endocrinol. 2018;14(8):452–63. actions in plasma and affected tissues in a diabetic
159. Ewan EE, et  al. Ascending dorsal column sensory mouse model. J Lipid Res. 2018;59(2):173–83.
neurons respond to spinal cord injury and down- 176. Szendroedi J, et  al. Role of diacylglycerol activa-
regulate genes related to lipid metabolism. Sci Rep. tion of PKCtheta in lipid-induced muscle insulin
2021;11(1):374. resistance in humans. Proc Natl Acad Sci U S A.
160. de Preux AS, et al. SREBP-1c expression in Schwann 2014;111(26):9597–602.
cells is affected by diabetes and nutritional status. 177. Chaurasia B, Summers SA.  Ceramides  - lipotoxic
Mol Cell Neurosci. 2007;35(4):525–34. inducers of metabolic disorders. Trends Endocrinol
161. Zhu L, et  al. Prolonged high-glucose exposure Metab. 2015;26(10):538–50.
decreased SREBP-1/FASN/ACC in Schwann cells 178. Hu W, et  al. Differential regulation of dihydrocer-
of diabetic mice via blocking PI3K/Akt pathway. J amide desaturase by palmitate versus monounsatu-
Cell Biochem. 2019;120(4):5777–89. rated fatty acids: implications for insulin resistance.
162. Tachibana H, et  al. Activation of liver X J Biol Chem. 2011;286(19):16596–605.
receptor inhibits osteopontin and amelio- 179. Fridman V, et  al. Randomized trial of l-serine in
rates diabetic nephropathy. J Am Soc Nephrol. patients with hereditary sensory and autonomic neu-
2012;23(11):1835–46. ropathy type 1. Neurology. 2019;92(4):e359–70.
Mechanisms of Nerve Injury in Diabetes: Dyslipidemia, Bioenergetics, and Oxidative Damage 305

180. Fridman V, et  al. Altered plasma serine and towards retinal capillary pericytes. J Lipid Res.
1-­deoxydihydroceramide profiles are associated 2014;55(5):860–9.
with diabetic neuropathy in type 2 diabetes and obe- 197. Nowicki M, et al. Oxidized low-density lipoprotein
sity. J Diabetes Complicat. 2021;35(4):107852. (oxLDL)-induced cell death in dorsal root ganglion
181. Rumora AE, et al. Plasma lipid metabolites associate cell cultures depends not on the lectin-like oxLDL
with diabetic polyneuropathy in a cohort with type 2 receptor-1 but on the toll-like receptor-4. J Neurosci
diabetes. Ann Clin Transl Neurol. 2021. https://doi. Res. 2010;88(2):403–12.
org/10.1002/acn3.51367. 198. Hinder LM, et al. Mitochondrial uncoupling has no
182. Afshinnia F, et  al. Increased lipogenesis and effect on microvascular complications in type 2 dia-
impaired beta-oxidation predict type 2 diabetic kid- betes. Sci Rep. 2019;9(1):881.
ney disease progression in American Indians. JCI 199. Eid SA, et al. Differential effects of empagliflozin
Insight. 2019;4(21):e130317. on microvascular complications in murine mod-
183. Stith JL, Velazquez FN, Obeid LM.  Advances in els of type 1 and type 2 diabetes. Biology.
determining signaling mechanisms of ceramide and 2020;9(11):347.
role in disease. J Lipid Res. 2019;60(5):913–8. 200. Hinder LM, et  al. Transcriptional networks of pro-
184. Raichur S, et al. The role of C16:0 ceramide in the gressive diabetic peripheral neuropathy in the db/db
development of obesity and type 2 diabetes: CerS6 mouse model of type 2 diabetes: an inflammatory
inhibition as a novel therapeutic approach. Mol story. Exp Neurol. 2018;305:33–43.
Metab. 2019;21:36–50. 201. Eid SA, et  al. Gene expression profiles of diabetic
185. Barber CN, Raben DM. Lipid metabolism crosstalk kidney disease and neuropathy in eNOS knockout
in the brain: Glia and neurons. Front Cell Neurosci. mice: predictors of pathology and RAS blockade
2019;13:212. effects. FASEB J. 2021;35(5):e21467.
186. Zlobine I, Gopal K, Ussher JR.  Lipotoxicity in 202. Sas KM, et al. Tissue-specific metabolic reprogram-
obesity and diabetes-related cardiac dysfunction. ming drives nutrient flux in diabetic complications.
Biochim Biophys Acta. 2016;1861(10):1555–68. JCI Insight. 2016;1(15):e86976.
187. McCoin CS, Knotts TA, Adams SH. Acylcarnitines— 203. Coppey L, et al. Effect of dietary oils on peripheral
old actors auditioning for new roles in metabolic neuropathy-related endpoints in dietary obese rats.
physiology. Nat Rev Endocrinol. 2015;11(10):617. Diabetes Metab Syndr Obes. 2018;11:117–27.
188. Hinder LM, et al. Comparative RNA-Seq transcrip- 204. Coppey L, et  al. Progressive loss of corneal nerve
tome analyses reveal distinct metabolic pathways in fibers and sensitivity in rats modeling obesity and
diabetic nerve and kidney disease. J Cell Mol Med. type 2 diabetes is reversible with omega-3 fatty acid
2017;21(9):2140–52. intervention: supporting cornea analyses as a marker
189. Guo K, et  al. Genome-wide profiling of DNA for peripheral neuropathy and treatment. Diabetes
methylation and gene expression identifies can- Metab Syndr Obes. 2020;13:1367–84.
didate genes for human diabetic neuropathy. Clin 205. Cinci L, et  al. Oxidative, metabolic, and apoptotic
Epigenetics. 2020;12(1):123. responses of Schwann cells to high glucose levels. J
190. Elzinga S, et al. Toll-like receptors and inflammation Biochem Mol Toxicol. 2015;29(6):274–9.
in metabolic neuropathy; a role in early versus late 206. Kato A, et  al. Recurrent short-term hypoglycemia
disease? Exp Neurol. 2019;320:112967. and hyperglycemia induce apoptosis and oxidative
191. Rocha DM, et al. Saturated fatty acids trigger TLR4-­ stress via the ER stress response in immortalized
mediated inflammatory response. Atherosclerosis. adult mouse Schwann (IMS32) cells. Neurosci Res.
2016;244:211–5. 2019;147:26–32.
192. Lee JY, et al. Saturated fatty acid activates but poly- 207. Liu YP, Shao SJ, Guo HD.  Schwann cells apopto-
unsaturated fatty acid inhibits Toll-like receptor sis is induced by high glucose in diabetic peripheral
2 dimerized with Toll-like receptor 6 or 1. J Biol neuropathy. Life Sci. 2020;248:117459.
Chem. 2004;279(17):16971–9. 208. Zhang L, et  al. Hyperglycemia alters the Schwann
193. Hwang DH, Kim JA, Lee JY.  Mechanisms for the cell mitochondrial proteome and decreases coupled
activation of Toll-like receptor 2/4 by saturated fatty respiration in the absence of superoxide production.
acids and inhibition by docosahexaenoic acid. Eur J J Proteome Res. 2010;9(1):458–71.
Pharmacol. 2016;785:24–35. 209. Russell JW, et al. Neurons undergo apoptosis in ani-
194. Stino AM, et  al. Evolving concepts on the role of mal and cell culture models of diabetes. Neurobiol
dyslipidemia, bioenergetics, and inflammation in the Dis. 1999;6(5):347–63.
pathogenesis and treatment of diabetic peripheral 210. Zilliox LA, Russell JW. Physical activity and dietary
neuropathy. J Peripher Nerv Syst. 2020;25(2):76–84. interventions in diabetic neuropathy: a systematic
195. Yokoi H, Yanagita M.  Targeting the fatty acid review. Clin Auton Res. 2019;29(4):443–55.
transport protein CD36, a class B scavenger recep- 211. Balducci S, et  al. Exercise training can modify the
tor, in the treatment of renal disease. Kidney Int. natural history of diabetic peripheral neuropathy. J
2016;89(4):740–2. Diabetes Complicat. 2006;20(4):216–23.
196. Fu D, et  al. Immune complex formation in human 212. Kluding PM, et  al. The effect of exercise on neu-
diabetic retina enhances toxicity of oxidized LDL ropathic symptoms, nerve function, and cutaneous
306 S. A. Eid et al.

innervation in people with diabetic peripheral neu- 224. Müller-Stich BP, et  al. Gastric bypass leads to
ropathy. J Diabetes Complicat. 2012;26(5):424–9. improvement of diabetic neuropathy independent
213. Singleton JR, et  al. Supervised exercise improves of glucose normalization–results of a prospec-
cutaneous reinnervation capacity in metabolic syn- tive cohort study (DiaSurg 1 study). Ann Surg.
drome patients. Ann Neurol. 2015;77(1):146–53. 2013;258(5):760–5; discussion 765–6
214. Smith AG, et  al. Lifestyle intervention for 225. Davis TM, et  al. Effects of fenofibrate on renal
pre-­diabetic neuropathy. Diabetes Care. function in patients with type 2 diabetes mel-
2006;29(6):1294–9. litus: the Fenofibrate Intervention and Event
215. Cooper MA, et  al. A ketogenic diet reduces meta- Lowering in Diabetes (FIELD) Study. Diabetologia.
bolic syndrome-induced allodynia and promotes 2011;54(2):280–90.
peripheral nerve growth in mice. Exp Neurol. 226. Keech AC, et  al. Effect of fenofibrate on the need
2018;306:149–57. for laser treatment for diabetic retinopathy (FIELD
216. Estruch R, et al. Primary prevention of cardiovascu- study): a randomised controlled trial. Lancet.
lar disease with a Mediterranean diet supplemented 2007;370(9600):1687–97.
with extra-virgin olive oil or nuts. N Engl J Med. 227. Rajamani K, et al. Effect of fenofibrate on amputation
2018;378(25):e34. events in people with type 2 diabetes mellitus (FIELD
217. Guasch-Ferré M, et al. Total and subtypes of dietary study): a prespecified analysis of a randomised con-
fat intake and risk of type 2 diabetes mellitus in the trolled trial. Lancet. 2009;373(9677):1780–8.
Prevención con Dieta Mediterránea (PREDIMED) 228. Cho YR, et al. Therapeutic effects of fenofibrate on
study. Am J Clin Nutr. 2017;105(3):723–35. diabetic peripheral neuropathy by improving endo-
218. Qian F, et al. Metabolic effects of monounsaturated thelial and neural survival in db/db mice. PLoS One.
fatty acid-enriched diets compared with carbohy- 2014;9(1):e83204.
drate or polyunsaturated fatty acid-enriched diets in 229. Kang EY, et  al. Association of statin therapy with
patients with type 2 diabetes: a systematic review prevention of vision-threatening diabetic retinopa-
and meta-analysis of randomized controlled trials. thy. JAMA Ophthalmol. 2019;137(4):363–71.
Diabetes Care. 2016;39(8):1448–57. 230. de Langen JJ, van Puijenbroek EP.  HMG-CoA-­
219. Lewis EJH, et al. Effect of omega-3 supplementation reductase inhibitors and neuropathy: reports to the
on neuropathy in type 1 diabetes: a 12-month pilot Netherlands Pharmacovigilance Centre. Neth J Med.
trial. Neurology. 2017;88(24):2294–301. 2006;64(9):334–8.
220. Wanders AJ, et  al. Fatty acid intake and its 231. Tierney EF, et  al. Association of statin use with
dietary sources in relation with markers of type peripheral neuropathy in the U.S. population 40
2 diabetes risk: The NEO study. Eur J Clin Nutr. years of age or older. J Diabetes. 2013;5(2):207–15.
2017;71(2):245–51. 232. Pergolizzi JV, et al. Statins and neuropathic pain: a
221. Bunner AE, et al. A dietary intervention for chronic narrative review. Pain Ther. 2020;9(1):97–111.
diabetic neuropathy pain: a randomized controlled 233. Kristensen FP, et al. Statin therapy and risk of poly-
pilot study. Nutr Diabetes. 2015;5:e158. neuropathy in type 2 diabetes: a Danish Cohort
222. The Look AHEAD Research Group. Effects of a Study. Diabetes Care. 2020;43(12):2945–52.
long-term lifestyle modification programme on 234. Oesterle A, Laufs U, Liao JK.  Pleiotropic effects
peripheral neuropathy in overweight or obese adults of statins on the cardiovascular system. Circ Res.
with type 2 diabetes: the Look AHEAD study. 2017;120(1):229–43.
Diabetologia. 2017;60(6):980–8. 235. Tsimihodimos V, Liberopoulos E, Elisaf
223. Diabetes Prevention Program Research Group. M.  Pleiotropic effects of fenofibrate. Curr Pharm
Long-term effects of lifestyle intervention or met- Des. 2009;15(5):517–28.
formin on diabetes development and microvascular 236. de Anda-Jauregui G, et al. Pathway crosstalk pertur-
complications over 15-year follow-up: the Diabetes bation network modeling for identification of con-
Prevention Program Outcomes Study. Lancet nectivity changes induced by diabetic neuropathy
Diabetes Endocrinol. 2015;3(11):866–75. and pioglitazone. BMC Syst Biol. 2019;13(1):1.
Targeting the Mitochondrion
in Diabetic Neuropathy

Ahmad Hedayat, Krish Chandrasekaran,
Lindsay A. Zilliox, and James W. Russell

1 Introduction mitochondrial function are encoded by nuclear


genes and are then manufactured in the cytosol,
Mitochondria are critical in regulating energy where specific targeting signals direct the pro-
homeostasis in both neurons and Schwann cells teins to mitochondrial surface receptors and then
in diabetic neuropathy. They synthesize ATP into specific mitochondrial compartments. Thus,
through oxidative phosphorylation but also have the proteins are very specifically directed to form
an important role in integrating cellular metabo- a highly complex molecular machinery in which
lism and cell signaling pathways. In addition to mitochondrial bioenergetics is only a small part.
the bioenergetic function of mitochondria in dia- Mitochondria have been shown to be highly
betes, research over the past 20 years shows that dynamic both in the cytosol and within the axon,
mitochondria have ever more complex roles and the mitochondria constantly undergo changes
(Fig.  1) in that mitochondrial bioenergetics is in morphology due to fission and fusion as part of
integrated with other metabolic pathways, pro- the process of mitophagy related to mitochon-
tein biogenesis, cellular signaling, quality con- drial quality control. These changes have been
trol, immune and stress responses, and apoptosis. shown to be important in diabetic neuropathy.
Importantly, 99% of the proteins involved in Furthermore, mitochondria are able to communi-
cate with other organ systems, for example, the
endoplasmic reticulum. Importantly, several
A. Hedayat · K. Chandrasekaran
Department of Neurology, University of Maryland studies have shown over the past 20 years that in
School of Medicine, Baltimore, MD, USA diabetic neuropathy there is loss of absolute
L. A. Zilliox mitochondria, a decrease in mitochondrial DNA,
Department of Neurology, University of Maryland and dysfunction of mitochondria that occurs at
School of Medicine, Baltimore, MD, USA multiple levels [1–6].
Maryland VA Healthcare System,
Baltimore, MD, USA
J. W. Russell (*)
Department of Neurology, University of Maryland
School of Medicine, Baltimore, MD, USA
Maryland VA Healthcare System,
Baltimore, MD, USA
CAMC Institute for Academic Medicine,
Charleston, WV, USA
e-mail: James.Russell@CAMC.org

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 307
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_17
308 A. Hedayat et al.

Fig. 1  Mitochondrial respiratory complexes, biogenesis, matrix side and provides CoQ for the respiratory chain
and quality control. Mitochondrial respiratory chain and enzymes. CoQ complex precursors are imported by
supercomplexes are integrated into functional networks the translocase of the outer membrane (TOM) and TIM
with the translocase of the inner membrane (TIM23) and machineries. The mitochondrial respiratory chain is the
the ATP-dependent AAA proteases of the inner mem- main generator for reactive oxygen species (ROS) that
brane. The ATP-dependent AAA proteases (not shown) increases oxidation of proteins, reduces protein synthesis,
act as a quality control system in mitochondria to degrade but can also has a role in signaling. CI complex I, CII
select inner membrane (IM), intermembrane space (IMS), complex II, CIII complex III, CIV complex IV, MPP mito-
matrix proteins, and outer membrane (OM) proteins. The chondrial processing peptidase, Oct1 octapeptidyl pepti-
coenzyme Q (CoQ) biosynthetic complex is on the IM dase, PAM presequence translocase-associated motor

2 Mitochondrial Mechanisms results in a high proton gradient, resulting in


and Potential Drug Targets increased reactive oxygen species (ROS) produc-
in DN tion. The increase in generation of ROS results
from mitochondrial dysfunction that is induced by
2.1 Role of Mitochondrial an increase in mitochondrial inner membrane
Dysfunction in DN depolarization and degeneration of mitochondria in
peripheral nerves [4, 6–10]. The regeneration of
Diabetes-induced-oxidative damage in neurons, new mitochondria may rescue these impaired mito-
axons, and Schwann cells has been proposed as a chondria. Thus, activation of mitochondrial regen-
unifying DN mechanism. An increased metabolic eration may be protective under conditions where
influx into mitochondria increases respiration and significant mitochondrial degeneration is present.
Targeting the Mitochondrion in Diabetic Neuropathy 309

Molecular methods allow manipulations of chondrial respiration proteins and protects and
the mouse genome to overexpress or knockout replicates the mitochondrial genome [3, 12, 15,
expression of individual genes. The ease of 16]. PGC-1α has been mapped to chromosome
genetic manipulation has provided important 4P15.1, a region associated with basal insulin
tools for study of the pathogenesis, prevention, levels in Pima Indians that are identified as hav-
and treatment of DN. There are three key genes, ing a high risk of developing diabetes and
which code for proteins that are transcriptional diabetic-­ related complications [17]. Common
factors or activators of transcriptional factors and polymorphisms of PGC-1α are associated with
promote function and regeneration of mitochon- conversion from impaired glucose tolerance to
dria (Fig.  2). These are: the NAD+-dependent diabetes [18]. Thus, it is likely that PGC-1α and
deacetylase SIRT1 (Silent mating type informa- its downstream signaling intermediates are
tion regulation 2 homolog1), Peroxisome important in normal mitochondrial regulation in
proliferator-­
activated receptor-gamma co-­diabetic subjects [19]. We used PGC-1α knock-
activator 1α (PGC-1α), and mitochondrial out mice [PGC-1α (−/−)] to study the effect in
Transcription Factor A (mtTFA or TFAM). DN. Loss of PGC-1α, a protein that is essential
Peroxisome proliferator-activated receptor-­ for mitochondrial function and energy homeosta-
gamma co-activator 1α (PGC-1α) is a transcrip- sis, causes neuropathy that is worsened by diabe-
tional co-activator and a master regulator for tes [4]. This observation was supported by
mitochondrial biogenesis in many tissues includ- evidence from other groups showing that phos-
ing the nervous system [11–13]. PGC-1α is a phorylation and expression of adenosine
promising target for therapy for neurological dis- monophosphate-­ activated protein kinase
ease. For example, the pan-PPAR agonist, bezafi- (AMPK)/PGC-1α and mitochondrial respiratory
brate, upregulates PGC-1α and exerts beneficial chain complex proteins are downregulated in
effects in a transgenic mouse model of dorsal root ganglia of both diabetic rats and mice
Huntington’s disease [14]. PGC-1α activates [6, 20, 21]. Control of bioenergetics in the dia-
transcriptional factors such as nuclear respiratory betic peripheral nerve is in part controlled by the
factor 1 (NRF1) and mitochondrial transcription SIRT1-AMPK and PGC-1α signaling axis in
factor A (TFAM), which in turn induces mito- which the upstream members (SIRT1-AMPK)

Fig. 2  The majority of mitochondrial proteins are synthe- system in response to increased metabolic loads provided
sized in the nucleus and shuttled to the mitochondria. Key by hyperglycemia or hyperlipidemia in diabetes. SIRT1
transcription factors such as SIRT1, PGC-1α, and TFAM regulates PGC-1α activity via deacetylation, whereas
are important in regulating mitochondrial function and PGC-1α regulates expression of TFAM via nuclear respi-
preserving mitochondrial DNA in the peripheral nervous ratory factor, NRF1
310 A. Hedayat et al.

sense the metabolic demands of cells, whereas drial activity in humans with insulin resistance
the downstream member, PGC-1α, regulates and diabetes [30, 31] and (2) previous studies
mitochondrial biogenesis and function [6, 20– showing PGC-1α knockout mice have degenerat-
23]. There was no difference in plasma glucose, ing mitochondria in the CNS [32].
hemoglobin A1C, or insulin levels between TFAM lies downstream of PGC-1α and has an
PGC-1α wild-type diabetic and PGC-1α knock- important role in protecting the integrity of mito-
out diabetic mice, indicating that exacerbation of chondria. In humanized TFAM overexpressing
peripheral nerve injury associated with reduced transgenic mouse model, levels of TFAM are
PGC-1α levels is independent of glycemic con- increased only twofold in DRG neurons [3].
trol. However, there was a significant increase in Overexpression of TFAM reversed experimental
total cholesterol and triglyceride levels in nondia- DN.  TFAM prevents slowing of nerve conduc-
betic PGC-1α knockout compared to PGC-1α tion velocity, decreases intra epidermal nerve
wild-type mice and PGC-1α knockout diabetic fibers’ loss, and reduces mechanical allodynia.
compared to PGC-1α wild-type diabetic mice. The transgenic mice expressing human TFAM
These findings are consistent with current obser- protein elevated mitochondrial DNA (mtDNA)
vations that increased lipid levels are associated copy number and total TFAM levels to the same
with neuropathy [24] possibly by ox-LDL inter- extent. This suggests that the mtDNA copy num-
action with the receptor LOX-1 [25–27]. Impaired ber itself does not have influence upon TFAM
fatty acid oxidation is strongly correlated to gene expression. The suggested explanation is
impaired activation of PGC-1α [28] and adipose-­ that the interaction between mtDNA and TFAM
specific ablation of PGC-1α results in mitochon- is interrelated and that the existence of one aug-
drial dysfunction [29]. Thus, mitochondrial ments the stability of the other. This interaction is
PGC-1α has a critical role in lipid metabolism. possibly valuable from a regulatory point of view
Loss of PGC-1α may also worsen neuropathy since minor changes in mtDNA or TFAM protein
due to impaired antioxidant defense related to levels result in swift modification to preserve a
impaired mitochondrial function. Several lines of constant ideal ratio between TFAM and mtDNA
evidence support this idea: PGC-1α was mark- [33]. This is supported by the studies that mtDNA
edly reduced in diabetic DRG neurons; the mito- interacts with TFAM to wrap mtDNA to consti-
chondria in neurons from PGC-1α nondiabetic tute a nucleoid formation similar to histones in
mice contained abnormal vacuoles consistent the nucleosome [34–36].
with mitochondrial degeneration; there was a Meanwhile, mitochondria can generate ROS
decrease in mitochondrial respiratory function; due to their respiratory activity, contingently
gene expression of glutathione peroxidase and TFAM functions in multiple roles to promote
SOD2 were reduced; protein levels of transcrip- mtDNA transcription, replication, and wraps
tion factors that regulate mitochondrial transcrip- mtDNA to protect it from ROS attack [34–36].
tion and are important for normal mitochondrial The blood studies demonstrated that there was no
homeostasis (for example, TFAM and NRF1) difference in glucose or insulin levels between
were significantly decreased in dorsal root gan- WT diabetic and TFAM transgenic diabetic mice.
glion neurons of both PGC-1α knockout nondia- Although human TFAM is likely to be expressed
betic and diabetic mice; levels of oxidized in all tissues because the promoter is not tissue-
proteins consistent with increased oxidative or cell-specific, TFAM overexpression does not
stress were significantly increased; and overex- reduce the severity of diabetes. Thus, protection
pression of PGC-1α, using an adenoviral con- against peripheral nerve injury is localized to
struct, prevented the generation of ROS in adult peripheral nervous system mitochondria and is
mouse DRG neuron cultures [4]. The role of independent of glycemic control. The results
PGC-1α in mitochondrial regulation are sup- showed that there is a net loss of both mtDNA
ported by: (1) data showing an inverse correlation and TFAM in chronic experimental diabetes.
between muscle PGC-1α levels and mitochon- After acute diabetic exposure, there is an attempt
Targeting the Mitochondrion in Diabetic Neuropathy 311

to upregulate both TFAM and mtDNA, whereas reverse neuropathy in a model of type 2 diabetes
the increase is not significant. It could be argued mellitus (T2DM). SIRT1 requires NAD+ as a
that exposure to acute hyperglycemia rises TFAM cofactor to deacetylate PGC-1α and other tran-
levels, mtDNA, and mitochondrial biogenesis to scriptional factors. Treatment with a precursor of
meet the high-energy demand in neurons [37]. NAD+, Nicotinamide Riboside (NR), can reverse
However, in chronic hyperglycemia with a con- neuropathy in the HFD mouse that is a model of
current generation of ROS, the regulation might T2DM [40–42]. Adult C57BL6 mice were fed a
change from physiological to pathological and high-fat diet (HFD, 60% calories from fat) for 2
could eventually lead to a decline in mtDNA, months until they developed neuropathy. Then,
TFAM, and mitochondrial function. MtDNA is 150 mg/kg or 300 mg/kg NR was mixed with
particularly susceptible to oxidative injury due in HFD and fed every day for 2 months [40, 41].
part to the following factors: first, its location Neuropathy improved with treatment in both
within mitochondria where the respiratory com- groups but more effectively with 300 mg/kg
plexes I and III are potential sites for the genera- NR. There was no change in the control diet ani-
tion of superoxide radicals and second, the mals. NR treatment decreased the HFD-induced
limited repair activity against DNA damage increase in triglycerides and non-esterified fatty
within mitochondria [38]. Under normal condi- acids and normalized the impaired glucose toler-
tions, the toxic effects of ROS are prevented by ance test. In HFD mice, there was a decrease in
such scavenging enzymes as superoxide dis- the NAD+ level, in SIRT1 activity, and in PGC-1α
mutase, glutathione peroxidase, catalase, and levels in DRG neurons. NR normalized these
other non-enzymatic antioxidants. However, measurements.
when ROS production becomes excessive, or if
the levels of antioxidant enzymes decrease, then
oxidative stress might harm the functional and 2.2 Changes in Redox Potential
structural integrity of biological tissue. DRG in the Mitochondrion in DN
neurons from TFAM transgenic mice are able to
scavenge high glucose-induced ROS much more Oxidative stress is one of the most important
efficiently than those from WT mice. However, potential mechanisms of injury to the peripheral
this scavenging ability did not appear to be due nervous system and most is generated from the
an increase in the expression of the antioxidant mitochondrion [7, 43]. Mitochondrial stress is
enzymes SOD2 or glutathione peroxidase. Our particularly important in T2DM but to a lesser
results with mitochondrial respiration showed no extent is important in type 1 diabetes mellitus
significant increase in ADP-stimulated State 3 (T1DM) [3, 4, 6, 23]. Stress states and dysfunc-
and resting State 4 respiration. All these findings tion of the respiratory chain can lead to increased
suggest that the protective effect of TFAM over reactive oxygen species (ROS) production that
expression is apparent only under chronic condi- causes oxidative damage in proteins, DNA, and
tions of oxidative stress. membranes. As ROS also have a signaling role,
SIRT1 acts upstream to coordinate the SIRT1/ mitochondrially produced ROS can signal the
PGC-1α/TFAM pathway to regulate mitochon- functional state of mitochondria [44–47]. In
drial oxidative energy metabolism and neuronal experimental diabetes, oxidative stress levels
protection. Using an inducible neuron-specific and reduced antioxidant defense are associated
SIRT1 overexpressing (SIRT1OE) C57BL6 with neuropathy. In comparison, blocking oxi-
mouse, SIRT1 is able to reverse DN in high fat dative stress in diabetic animals re-establishes
fed diabetic mice [39]. Diabetes reduces NAD+, normal blood flow and sciatic and saphenous
SIRT1, and PGC-1α levels in DRG neurons. nerve conduction velocities. Augmented meta-
SIRT1 overexpression normalizes these levels. bolic mitochondrial flux due to high glucose
These findings are consistent with the concept results in increased reactive oxygen species
that the SIRT1/PGC-1α/TFAM pathway can (ROS) formation such as superoxide, peroxyni-
312 A. Hedayat et al.

trite, hydroxyl radicals, and impaired mitochon- normal conditions, Nrf2 is inactive and remains
drial respiratory function [21]. ROS generation in the cytosol. Acute hyperglycemia increases
enhances mitochondrial permeability transition, the expression of Nrf2, but persistent hypergly-
opening the adenine nucleotide translocase/ cemia decreases Nrf2 expression.
voltage-­dependent anion channel resulting in Downregulation of Nrf2 causes various micro-
mitochondrial swelling that distorts the outer vascular changes that result in diabetic neuropa-
membrane’s integrity [1, 4]. The resulting mem- thy. Targeting Nrf2 activators as potential
brane lipid peroxidation and DNA degradation therapies may provide insight into new therapies
are linked to neuronal or axonal injury [48]. for diabetic neuropathy. For example, natural
Mitochondria become swollen and develop dis- compounds such as curcumin, sulforaphane, res-
rupted cristae [48], and there is loss of mito- veratrol, and vitamin D can activate Nrf2 and
chondria [4]. promote antioxidant pathways to mitigate oxida-
In contrast, upregulation of pathways that tive stress [52]. Another factor that can trigger
block oxidative stress precludes axonal injury in Nrf2 is taurine, found naturally in the body.
diabetic neuropathy. For example, overexpres- Supplementing diabetic rats with taurine did not
sion of manganese superoxide dismutase (SOD2), alter body weight and blood glucose concentra-
the enzyme responsible for mitochondrial detoxi- tion, but reduced the serum malondialdehyde
fication of oxygen radicals, reduces superoxide in concentration in diabetic rats and alleviated neu-
cultured dorsal root ganglion (DRG) neurons and roinflammation by suppressing NF-kappaB
inhibits cellular injury. Diabetic neuropathy is expression and enhancing Nrf2 [53].
observed in the C57BL/6Jdb/db diabetic mouse, Fisetin, a phytoflavonoid that simultaneously
and decreased expression of SOD2 in these ani- targets NF-kappaB and Nrf2, shows a beneficial
mals increased diabetic neuropathy [49]. The effect in experimental DN [54]. Fisetin was
generation of ROS can be decreased by stabiliz- administered at 5 and 10 mg/kg for 2 weeks in
ing the inner mitochondrial membrane potential streptozotocin (STZ) diabetic rats. As deter-
and reducing generation of superoxide and other mined by nerve conduction studies and sciatic
radicals or by increasing antioxidant defense. nerve blood flow deficits, neuropathy was ame-
Rising levels of uncoupling proteins such as liorated in treated rats. Importantly, fisetin
UCP1 or UCP3 that prevent hyper-polarization reduced levels of interleukin-6 and tumor necro-
of the inner mitochondrial membrane potential sis factor-alpha in sciatic nerves of diabetic rats
will reduce ROS formation and ameliorate neuro- (p < 0.001), suggesting that a combined approach
nal injury [1]. to modifying oxidative stress and neuroinflam-
An important marker of oxidative stress is a mation may be a more effective approach. The
reduction in the glutathione system that protects mechanistic target of rapamycin (mTOR) is
diabetic neurons and Schwann cells from injury. another promising agent for the development of
Diabetic patients with distal symmetric polyneu- novel regenerative strategies for the treatment of
ropathy exhibited significantly lower values of diabetes mellitus. The mTOR pathway impacts
reduced glutathione (GSH) and a reduced gluta- numerous cellular processes including cellular
thione/oxidized glutathione (GSH/GSSG) ratio metabolic homeostasis, insulin resistance, insu-
[50]. In contrast, malondialdehyde and oxidized-­ lin secretion, apoptosis, and autophagy [55].
LDL levels were unchanged in diabetic neuropa- mTOR is critical for many signaling pathways
thy. Another recent approach has been to that involve phosphoinositide 3-kinase (PI 3-K),
manipulate Nrf2, which mediates various anti- protein kinase B (Akt), AMP-­activated protein
oxidant proteins’ expression via antioxidant kinase (AMPK), sirtuin 1 (SIRT1), Wnt1 induc-
response element (ARE) binding sites. Nrf2 ible signaling pathway protein 1 (WISP1), and
appears to bridge the link between neuroinflam- insulin/insulin growth factor 1 [56]. mTOR, sir-
mation and apoptotic pathways impacting the tuins, and insulin/insulin growth factor 1 signal-
progression of diabetic neuropathy [51]. Under ing are longevity pathways involved in an array
Targeting the Mitochondrion in Diabetic Neuropathy 313

of different processes, including metabolism, peripheral nervous tissue [65]. Tissue glucose is
and neuronal plasticity, that are critical in pre- metabolized to 3-deoxyglucosone (3-DG),
venting neuropathy and in axonal repair. As a methylglyoxal, and Nε-(carboxymethyl) lysine.
result, mTOR represents an exciting target for Binding of these AGE ligands to the receptor for
the treatment of DN. advanced glycation end-products (RAGE) can
Nitrosative stress can increase nerve injury in result in diabetic complications. AGE and
diabetes [57]. Peroxynitrite resulting from the RAGE accumulation in nerve perineurial, epi-
reaction of nitric oxide with superoxide causes neurial, and endoneurial microvessels of
oxidative stress and leads to poly ADP-­ patients with impaired glucose tolerance (IGT)
ribosylation, mitochondrial dysfunction, results in increased expression of the transcrip-
impaired stress signaling, and protein nitration. tion factor nuclear factor κB (NFκB) [66, 67].
In diabetic mice treated with the peroxynitrite AGE:RAGE interactions further lead to the gen-
decomposition catalyst Fe(III) tetramesitylpor- eration of ROS (ROS) and the activation of pro-
phyrin octasulfonate (FeTMPS, 10 mg/kg/day) tein kinase C (PKC), transforming growth factor
or protein nitration inhibitor(-)-epicatechin gal- β (TGF-β) [68], mitogen-activated protein
late (20 mg/kg/day) for 4 weeks, there was partial kinase (MAPK), and transcription factors like
correction of neuropathy [58]. This supports the nuclear factor κB (NFκB) [69].
concept that peroxynitrite decomposition cata- RAGE is thought to be one of the potential
lysts may be used in the treatment of diabetic contributors to neurotoxicity [70]. It has been
peripheral neuropathy. In humans, gastric bypass shown that RAGE activation leads to an increase
and in particular Roux-Y gastric bypass (RYGB) in proinflammatory molecules, oxidative stress-
can improve neuropathic symptoms independent ors, and cytokines. The proinflammatory effect of
from glycemic control. The procedure is associ- RAGE is mediated by its binding to ligands such
ated with improved nitrosative injury [59], and as AGE, S100/calgranulin, and amphoterin.
the decrease in nitrotyrosine is correlated with Activation of these ligands leads to subsequent
improvement in the neuropathy disability scale activation of downstream pathways such as
up to 12 months. NF-kappaB, STAT, and JKN. An important target
for the treatment of AGE-induced diabetic nerve
injury is the glyoxalase system. The glyoxalase
2.3 Advanced Glycation End system is a highly specific enzyme system exist-
Products ing in all mammalian cells that is responsible for
and the Mitochondrion in DN the detoxification of dicarbonyl species, primar-
ily methylglyoxal. It has been implicated to play
Dicarbonyl metabolites, for example methylgly- an essential role in preventing the increased for-
oxal, glyoxal and 3-deoxyglucosone, accumula- mation of AGEs. When glyoxalase 1 is knocked
tion has been associated with metabolic and out in cells, the formation of methylglyoxal and
age-related diseases in which there is a pro-­ associated protein modifications were blocked
inflammatory and pro-oxidant state [60]. [71], suggesting that glyoxalase 1 could be an
Methylglyoxal is the most reactive dicarbonyl important target for treatment in DN [72]. Other
and is the precursor of the major quantitative studies have shown that pyridoxamine prevents
advanced glycation end products (AGEs). the formation of AGEs by trapping carbonyl
Improved mitochondrial function has been intermediates and can reduce development of
linked to a decrease in generation of AGEs in a vascular disease and nephropathy in the nondia-
variety of tissues [61], and specifically in rela- betic, Zucker obese rat [73], but little is known
tion to diabetic neuropathy and other diabetic about its effect in reducing human diabetic neu-
complications [62–64]. In contrast, increased ropathy in human studies. Other natural inhibi-
glycemia results in reduced mitochondrial func- tors of AGEs have not been tested in diabetic
tion and induces the generation of AGEs in neuropathy.
314 A. Hedayat et al.

2.4 Mitochondrial Regulation nephropathy and retinopathy because of its abil-


of Molecular Chaperones ity to increase extracellular matrix.
in DN There is evidence that TGF-β is upregulated in
STZ diabetic rats with neuropathy. In DRG and
Heat shock proteins (HSPs) or molecular chaper- sciatic nerve from diabetic animals, TGF-beta1
ones also have roles in cell signaling and regula- and TGF-beta2 are increased and are associated
tion of metabolism. Overexpression and with increased neuronal death [68]. In diabetic
pharmacological induction of HSP72 prevent DRG neurons using quantitative real-time PCR
high-fat diet-induced glucose intolerance and (QRT-PCR), TGF-β1 and TGF-β2 mRNA, but
skeletal muscle insulin resistance [74]. not TGF-β3, were increased at the 4 and 12 week
Furthermore, overexpression of skeletal muscle time point [68]. In the sciatic nerve, TGF-β3
HSP72 in mice increases endurance nearly two- mRNA was primarily increased. In DRG neu-
fold and increases mitochondrial content by 50%. rons, pretreatment with TGF-β neutralizing anti-
HSP72 mRNA expression correlates well with body prevented the increase in total TGF-β
mitochondrial enzyme activity in human skeletal protein observed with high glucose. In DRG neu-
muscle. In contrast, HSP72 expression is mark- rons exposed to high glucose, TGF-β2 > β1
edly decreased in skeletal muscle in humans with increased the percent of cleaved caspase-3 com-
type 2 diabetes. pared to high glucose alone, and TGF-β neutral-
Individual molecular chaperones and chaper- izing antibody inhibited the increase. TGF-β
one complexes are expressed under normal and isoforms applied directly to DRG neurons
disease states. Molecular chaperones like heat reduced neurite outgrowth, and this effect was
shock protein 90 (Hsp90) and Hsp70 assist in partially reversed by TGF-β neutralizing anti-
folding nascent polypeptides into their final bio- body. These findings implicated upregulation of
logically active conformations and in the refold- TGF-beta in experimental diabetic peripheral
ing of aggregated and denatured proteins. As a neuropathy and suggested a potential new target
result, the chaperones help direct proteins toward for the treatment of diabetic peripheral
degradation via the proteasome or by autophagy neuropathy.
[75, 76]. Pharmacologic modulation of the Patients with T2DM and clinically detectable
molecular chaperones can improve insulin resis- serum TGF-β1 showed a positive correlation with
tance [77] and peripheral neuropathy [78]. Hsp90 nerve conduction velocities, suggesting that this
modulators such as DMAG, KU-32, and KU-596 cytokine might be used as a biomarker for dia-
show promise in treating diabetic complications betic peripheral neuropathy [81]. TGF-β increases
in animal models and may afford a novel and Smad3 signaling, thereby affecting metabolism
effective disease-modifying approach for humans and energy homeostasis and reducing inflamma-
[79]. tion and ROS production [82].

2.5 TGF-β and DN 2.6 Metabotropic Glutamate


Receptors, Oxidative Injury,
Transforming growth factor-β1 (TGF-β1) is an and DN
important mediator of fibrosis in diabetes. Data
from human biopsy material and experimental Metabotropic glutamate receptors (mGluRs) are
models indicate increased expression of IHG-1 is a subfamily of glutamate receptors that are
a critical component of fibrogenesis as it ampli- G-protein-coupled and linked to second messen-
fies TGF-β1 signaling and biogenesis [80]. IHG-1 ger systems [83]. Glutamate is the principal neu-
is expressed in mitochondria, stabilizes PGC-1α rotransmitter in the CNS that is transported to the
protein, and increases mitochondrial biogenesis. axon terminals [84]. The mechanism for produc-
TGF-β1 has been associated with diabetic tion, paracrine release, and recycling of gluta-
Targeting the Mitochondrion in Diabetic Neuropathy 315

mate happens in sensory ganglia and contains the of DN in animal models [89]. mGluR2/3 ago-
enzymes amidohydrolase, glutaminase [85, 86], nists prevent glucose-induced neuronal injury in
glutamate aspartate transporter (GLAST), gluta- DRG neuronal cultures by increasing glutathi-
mate transporter 1 (GLT1) [87], as well as the one and maintaining mitochondrial function
recycling enzyme glutamine synthetase [85, 88]. only in the presence of Schwann/SGC cells [83,
The Glutamate carboxypeptidase II (GCP II) 90, 91]. Treatment with a mGluR2/3 agonist nor-
inhibitor 2-(phosphonomethyl)pentanedioic acid malizes levels of glutathione and oxidized pro-
(2-PMPA), which activates mGluR2/3, protects teins, while increasing levels of superoxide
against glucose-induced programmed cell death dismutase 2 (SOD2), SIRT1, PGC-1alpha,
(PCD) and neurite degeneration in DRG neurons TFAM, glutamate transporter proteins, and glu-
in a cell culture model of DN [83]. Preclinical tamine synthetase in DRG neurons. Thus,
data indicate that GCPII inhibitors ameliorate mGluR3 agonists have the ability to protect
DN in animal models. Direct or indirect activa- against cellular injury by regulating oxidative
tion of mGluR2/3 protects against development stress in models of DN (Fig. 3).

Fig. 3  Glutamate transporters GLT-1 and GLAST are lular glutamate. LY379268 treatment promotes glutamate
present in Satellite Glial Cells (SGC) cells to transport uptake, and it likely decreases extracellular glutamate. In
extracellular glutamate into the SGC where glutamine addition, mGluR 2/3 receptors present in DRG neurons
synthetase (GS) converts it to glutamine, which is eventu- decrease extracellular glutamate. Thus, activation of
ally recycled to the neuron for conversion into glutamate. mGluR2/3 receptor by an agonist can likely constrain sen-
In diabetes, hyperglycemia-induced oxidative stress sory transmission and importantly reduce nociceptive
affects glutamate transport proteins, increasing extracel- transmission
316 A. Hedayat et al.

2.7 Mitophagy, plement of DNA in the mitochondria presents a


the Inflammasome, range of challenges to the host immune system
and Innate Immune Pathways [104]. Release of mtDNA has been shown to trig-
in DN ger DNA-sensing antiviral pathways [104].
Activation of the innate immune system occurs in
Recent results suggest that mitochondrial impair- peripheral neuropathy and is of importance. For
ment contributes to inflammasome formation and example, vincristine-induced peripheral neuropa-
activation of the innate immune system. thy is driven by activation of the NLRP3 inflam-
Maintaining functional mitochondria is essential masome and subsequent release of interleukin-1β
to generate energy (ATP) for maintaining resting from macrophages. However, if inflammation is
potentials, action potentials, and neurotransmis- reduced by treatment with the NLRP3 inhibitor
sion at both the pre- and post-synaptic sites in the MCC950, or if there is knockout of NLRP3  in
neuron [92, 93]. Mitochondria, therefore, need to mice, then there is a significant reduction in
be repaired and replaced [94]. Mitochondrial bio- mechanical allodynia and gait impairment [105,
genesis, axonal transport, and mitochondrial fis- 106]. Similarly, paclitaxel can induce high-level
sion and fusion contribute to this rejuvenation expression of NLRP3 inflammasomes in the
[95]. Mitochondrial stress has been proposed as a infiltrated macrophages within DRG and sciatic
major mediator of diabetic complications such as nerve. This in turn promotes IL-1β production
neuropathy in humans and animal models [7, 43, and mechanical allodynia in a chemotherapy-­
96]. Therefore, in order to maintain a healthy induced neuropathic pain model [107].
mitochondrial population, damaged proteins and The NF-kappaB pathway is part of the central
organelles must be cleared. Mitochondria machinery initiating and propagating inflamma-
undergo a process of regeneration and autophagy tory responses, and this is true also for DN. The
(clearance), termed mitophagy. Impaired mito- NF-kappaB inflammatory cascade may be inhib-
chondrial quality control can cause disease, for ited by BAY 11-7082, an IkappaB phosphoryla-
example, mitofusin 2 mutations in Charcot-Marie tion inhibitor [108]. BAY 11-7082 (1 and 3 mg/
Tooth disease type 2A [97, 98] and Opa1 muta- kg) was administered to STZ diabetic rats for 14
tions in autosomal-dominant optic atrophy [99– days starting from the end of the 6th weeks post
101]. In addition, impairment of mitochondrial diabetic induction. Diabetic rats developed
quality control has been demonstrated to activate altered nociceptive parameters and deficits in
innate immune pathways, including inflamma- nerve functions and also demonstrated elevated
some (NLRP3)-mediated signaling and the anti- expression of NF-kappaB (p65), IkappaB, and
viral cyclic GMPAMP synthase (cGAS)/ p-IkappaB along with elevated levels of IL-6 and
stimulator of interferon genes (STING)–regu- TNF-alpha and inducible enzymes (COX-2 and
lated interferon response [reviewed in [95]]. iNOS). In addition, DN was associated with a
Innate immune-signaling pathways have evolved surge in oxidative stress, a reduction in Nrf2/
in complex multicellular eukaryotes to recognize HO-1 expression, and an amelioration in GSH
invasive pathogens, including bacteria, viruses, levels. BAY 11-7082 improved abnormal sensory
fungi, and protists. Damage-associated mito- responses and deficits. BAY 11-7082 also facili-
chondrial protein (DAMP) is recognized by the tated the surge in the expression of NF-kappaB,
cell’s immune-recognition pathways as foreign IkappaB, and p-IkappaB. BAY 11-7082 restrained
due to the resemblance to ancient prokaryotes the levels of IL-6, TNF-alpha, COX-2, and iNOS
[102]. Sensing of these DAMPs by NOD-, LRR-, in the sciatic nerve. Thus, it can be concluded that
and pyrin domain–containing proteins (NLRPs) NF-kappaB expression and downstream expres-
has been shown to activate inflammatory immune sion of proinflammatory mediators are significant
responses [103]. Like the problems posed by features of nerve damage resulting in inflamma-
mitochondria’s resemblance to ancient prokary- tion and oxidative stress. Furthermore, BAY
otes, the presence of a second nonnuclear com- 11-7082 is able to improve experimental DN by
Targeting the Mitochondrion in Diabetic Neuropathy 317

modifying neuroinflammation and improving deprivation leads to an increase in NAD+ and a


antioxidant protection. decrease in nicotinamide (NAM), which in turn
leads to activation of SIRT1 and 35 kDa peroxi-
some proliferator-activated receptor-gamma
3 Current Treatment co-activator-1α (PGC1-α) in the mitochondrion,
Approaches That Target resulting in an increase in oxidative phosphoryla-
Mitochondrial Function tion and fatty acid metabolism [2, 39, 109, 113].
in DN NAM and NADH act as inhibitors of SIRT1.
NAM phosphoribosyltransferase (Nampt) is
3.1 Lifestyle Modification important in the conversion of NAM to NAD+.
and Improvement Exercise increases Nampt activity in muscle but
in Mitochondrial Function also circulates outside the muscle to increase
in DN insulin sensitivity. Nampt then decreases NAM
and increases NAD+ resulting in further activa-
There are currently no disease-modifying treat- tion of the SIRT1-PGC1-α pathway and increas-
ments that have been definitively shown, in ing mitochondrial function. AMP-activated
­randomized clinical trials, to reduce or reverse protein kinase (AMPK) also likely regulates
diabetic sensory polyneuropathy. However, indi- SIRT1 by acting as an energy sensor and acti-
viduals with impaired glucose regulation and vates Nampt. Thus, the net effect of nutritional
neuropathy may benefit from aggressive diabetic restriction and exercise is to increase NAD+ and
control and lifestyle interventions. These inter- drive the SIRT1-PGC1-α pathway toward
ventions can postpone the onset of diabetes and improved mitochondrial function.
may reverse small fiber neuropathy associated In addition, changes in fatty acid metabolism
with early diabetes mellitus. Exercise not only likely affect mitochondrial function in the mus-
increases insulin sensitivity and glucose control, cle. Translational research indicates that high-fat
but also increases end organ perfusion, reduces diets can result in neuropathy in diabetes and that
lipid and protein oxidation, inhibits adipocyte withdrawing or otherwise manipulating a high fat
production of free fatty acids and deleterious adi- diet can reduce neuropathy [114–116]. In
pokines, and reduces humoral inflammation [1, 9, humans, a nutritional intervention study that was
109–111]. The mechanisms by which lifestyle targeted to improve essential fatty acid dys-
changes can affect mitochondrial function have metabolism in type 1 diabetes mellitus examined
previously been described [109]. Although supplementation with seal oil omega-3 polyun-
dietary and lifestyle interventions are likely to saturated fatty acids in individuals with type 1
affect multiple pathways, there is evidence that diabetes mellitus and neuropathy. After 12
improved mitochondrial function in muscle months of supplementation, there was an increase
might exert a remote effect. AMP-activated pro- in corneal nerve fiber length [117]. There was no
tein kinase (AMPK) and the protein deacetylase, progression of clinical disease symptoms, and
sirtuin 1 (SIRT1), are fuel sensing molecules. there were no declines in small and large fiber
During energy deprivation, AMPK activation sensory and functional measures. However, there
restores energy balance by increasing levels of was no improvement detected in nerve conduc-
ATP, for example by increasing fatty acid oxida- tion studies or sensory function.
tion, and reducing processes that consume
ATP.  AMP-activated protein kinase has been
shown to circulate in the blood after release from 3.2 Improved Glycemic Control
muscle [112]. This could explain why exercise, and DN
which improves muscle function, can also have
remote effects on somatic and autonomic nerve As discussed above, improved glycemic control
fibers. Furthermore, in exercised muscle, nutrient will reduce mitochondrial dysfunction and oxida-
318 A. Hedayat et al.

tive or nitrosative stress, consequently reducing rate of hypoglycemic complications, particularly


protein and lipid oxidation, neuroinflammation, in the insulin treatment group [120].
and neuronal and Schwann cell injury signaling. Other studies have shown a less definitive
Early hyperglycemic insult can lead to perma- change in neuropathy in T2DM with improved
nent, cumulative damage. Mitochondrial injury glycemic control. For example, the VA Diabetes
takes place after just 1 h of hyperglycemic expo- Trial randomized 1791 military veterans with
sure due to an increase in mitochondrial mem- T2DM to either intensive or standard glucose
brane potential likely caused by a decrease in the control. The development of neuropathy was
cells’ NAD+/NADH ratio [118]. The Diabetes determined based upon self-report. While the
Control and Complications Trial (DCCT) deter- study did not find any difference in the rate of
mined a clear link between impaired glycemic new microvascular complications, such as neu-
control, neuropathy, and retinopathy. This DCCT ropathy, there was a non-significant 5% reduction
prospectively followed 1441 participants with in the incidence of neuropathy [121]. Not all
T1DM for a mean of 6.5 years to study the effect studies have shown that aggressive glucose con-
of intensive insulin on the development of dia- trol reduces the development or progression of
betic complications [reviewed in [111]]. The neuropathy in T2DM. The VA cooperative study
study divided participants into a primary (preven- on type II diabetes mellitus (VA-CSDM) com-
tion) and a secondary (intervention) group and pared standard insulin treatment to an intensive
treated them with intensive or conventional insu- therapy group in men with T2DM who required
lin therapy. In the secondary-intervention cohort, insulin. The enrolled men had poorly controlled
intensive insulin therapy reduced clinical neu- T2DM with an average duration 7.8 ± 4 years and
ropathy’s appearance by 60% at the 5-year fol- about half had neuropathy at baseline. Intensive
low-­up. If patients had neither retinopathy nor insulin therapy did result in an improvement in
significant albuminuria at the start of the study HgBA1c after 6 months, but no difference was
(primary-prevention cohort), then the results found in the prevalence of DN between the inten-
were even more impressive: intensive therapy sive vs. standard arms (64% intensive vs 69%
reduced the appearance of neuropathy by 69% standard) after 2 years [122].
compared to only 10% with conventional therapy Importantly, improved glycemic control has
and showed that early optimal glycemic control been shown to have a sustained benefit on diabe-
could prevent the development of neuropathy tes and its complications. The NeuroEDIC study
before patients developed microvascular injury found a reduced prevalence of neuropathy in a
and retinopathy. group of type 1 diabetics that received intensive
Although studies show clear improvement in treatment compared to the standard treatment
diabetic and somatic neuropathy outcomes for group during the DCCT study. However, although
T1DM, the data for T2DM is less clear [119]. intensive glycemic control reduced the severity
Despite these reservations, improvement in of neuropathy, 34% of subjects in the former
T2DM was observed in the UK Prospective intensive treatment group and 41% of those in the
Diabetes Study (UKPDS) study. In this study, former conventional treatment group still devel-
3867 patients with newly diagnosed T2DM were oped clinical neuropathy [123].
randomly assigned to intensive therapy with a
sulfonylurea (chlorpropamide, glibenclamide, or
glipizide), with insulin, or conventional diet ther- 3.3 Diet and Lifestyle
apy. After 10 years, risk of microvascular end- Interventions in DN
points was reduced by 25% (p = 0.0099) [120].
Thus, intensive blood glucose control by either Glucose-modifying drug therapy is typically not
sulfonylureas or insulin substantially decreased appropriate in patients with impaired glucose
the risk of microvascular complications in regulation due to cost and potential for serious
patients with T2DM; however, there was a higher side effects such as hypoglycemia. A more suit-
Targeting the Mitochondrion in Diabetic Neuropathy 319

able approach for these patients would be a life- drug intervention in preventing conversion from
style intervention that could arrest the underlying IGT to diabetes [124]. Furthermore, studies have
process that leads to neuropathy and its associ- shown that an exercise intervention can prevent
ated functional disability. At present there is no the development of neuropathy in subjects who
evidence from a randomized study that a lifestyle have T2DM but do not have neuropathy [127–
intervention would reverse somatic neuropathy. 129], may affect pain and symptom outcomes in
However, there is evidence that a lifestyle inter- subjects with neuropathy, and is safe [130, 131].
vention can be more effective than a drug inter-
vention in preventing conversion from IGT to
diabetes. The Diabetes Prevention Program 3.4 Alpha-Lipoic Acid and DN
(DPP) study was designed to determine if a more
intensive dietary and exercise intervention or Alpha-lipoic acid (ALA) is biosynthesized in
metformin treatment effectively prevented or small amounts from octanoic acid in the mito-
delayed the onset of T2DM in people with IGT or chondrion, where it is used as a cofactor for pyru-
impaired fasting glucose (IFG). The DPP demon- vate dehydrogenase and α-ketoglutarate
strated that lifestyle changes reduced the risk of dehydrogenase complexes [132, 133]. ALA is a
developing T2DM by 58% in adults with IFG or strong antioxidant that functions by removing
IGT who were at high risk of developing diabetes already formed free radicals [134]. ALA has a
[124]. Metformin drug therapy did also reduce disease modifying effect, and in several studies, it
the risk of developing T2DM, but was less effec- has been shown to improve DN symptoms.
tive than weight loss and increased physical However, the evidence to support its use is not
activity [124]. To prevent one case of diabetes in strong because clinical trials with ALA have been
3 years, only 6.9 persons would have to partici- completed using a variety of study designs, routes
pate in the lifestyle intervention program, of administration, and sample sizes [135–138]. In
whereas 13.9 would have to receive metformin the multicenter, randomized, double-blind,
[124], which clearly showed that the lifestyle placebo-­controlled ALADIN III trial, there was a
intervention was almost twice as effective as drug small but significant improvement in the
therapy. Examining the 10-year cost-­effectiveness Neuropathy Impairment Score (NIS) in ALA-­
of the DPP interventions, lifestyle was found to treated patients, but no significant improvement
be cost-effective, and metformin was marginally in the Total Symptom Score (TSS) [136]. In the
cost-saving compared to placebo [125]. Deutsche Kardiale Autonome Neuropathie
Optimally, lifestyle changes should be intro- (DEKAN) Study, there were small improvements
duced in patients with IGT, who are early in the in the cardiac autonomic spectral analysis in
course of developing neuropathy. The Impaired ALA treated patients [139]. In the SYDNEY2
Glucose Tolerance Causes Neuropathy Study trial, 181 diabetic patients were treated for 5
(IGTN) was a natural history study that gave gen- weeks after a 1-week placebo run-in period with
eral dietary and physical activity advice (similar the following daily oral doses of ALA: 600 mg (n
to those in the DPP “lifestyle intervention” = 45) (ALA600), 1200 mg (n = 47) (ALA1200),
group) to participants with IGT or IFG and mild and 1800 mg (n = 46) (ALA1800) or placebo (n
neuropathy. In the IGTN study, weight loss and/ = 43) [137]. The change from the baseline TSS
or an increase in physical activity was associated was the primary outcome measure. The
with slower progression of neuropathy based on Neuropathy Symptoms and Change (NSC) score
the intraepidermal nerve fiber density and with and the NIS were secondary end points. The
the ability to re-grow epidermal nerve fibers mean TSS decreased by 51% in ALA600, 48% in
[110, 126]. More recent studies have underscored ALA1200, and 52% in ALA1800 compared with
the importance of intervention during the earliest 32% in the placebo group (P < 0.05 vs. placebo).
stages of impaired glucose regulation, and a life- The NSC significantly improved in all three ALA
style intervention can be more effective than a groups, and the NIS was numerically reduced.
320 A. Hedayat et al.

Thus, oral treatment with ALA for 5 weeks higher neuropathy stage. A more recent study
improved neuropathy measures in patients with used a combination therapy with superoxide dis-
DN. The optimum risk-to-benefit ratio occurred mutase, ALA, acetyl L-carnitine, and vitamin
with 600 mg alpha-lipoic acid once daily [137]. B12  in a prospective, double-blind, placebo-­
Further support for ALA therapy is provided by a controlled study, randomized study in 85 patients
meta-analysis of four trials (ALADIN I, ALADIN with T2DM over 12 months [141]. In this study,
III, SYDNEY, NATHAN II) comprising 1258 the Michigan Neuropathy Screening Instrument
patients (ALA n = 716; placebo n = 542) [140]. Questionnaire, vibration perception threshold,
In the Nathan I study [138], 460 diabetic patients sural nerve conduction velocity and amplitude,
with mild-to-moderate diabetic sensory polyneu- pain, and quality of life questionnaires were mea-
ropathy were assigned randomly to oral treat- sured endpoints and showed statistically signifi-
ment with 600 mg ALA once daily (n = 233) or cant improvement after 12 months compared to
placebo (n = 227) for 4 years. A composite score the placebo group. The cardiovascular autonomic
of the NIS-LL and seven neurophysiologic tests reflex tests and Michigan Neuropathy Screening
was the primary end point. The NIS, NIS-LL, Instrument Examination did not significantly
nerve conduction, and quantitative sensory tests change. However, similar results were obtained
(QSTs) were included as secondary outcome with normalization of just B12 levels [142] sug-
measures. There was no significant difference gesting that the main active component in the
between treatment groups for the change in pri- combination tablet was B12.
mary end point from baseline to 4 years (P =
0.105). When sub-scores of the composite score
were measured independently, the change from 3.5 Benfotiamine and DN
baseline was significantly better for ALA com-
pared to placebo for: NIS (P = 0.028), NIS-LL (P Mitochondria take up and use thiamine pyro-
= 0.05), and the NIS-LL muscular weakness sub-­ phosphate, the active form of thiamine. In dia-
score (P = 0.045). With ALA compared to pla- betic tissues, only about 10% of a cell’s thiamine
cebo, more patients showed a clinically is in the cytosol, available to transketolase, while
meaningful improvement, and fewer showed pro- the remainder is inside the mitochondrion [118].
gression in NIS (P = 0.013) and NIS-LL (P = Thiamine plays a crucial role in the metabolism
0.025). Part of the reason that the primary study of energy. Benfotiamine is a synthetic derivative
endpoint failed to detect a difference was that of thiamine. It elevates the levels of thiamine
nerve conduction and QST results did not signifi- diphosphate in the cell, a cofactor necessary for
cantly worsen in the placebo group. Global the activation of transketolase, resulting in the
assessment of treatment tolerability did not differ reduction of tissue levels of AGEs. The anti-AGE
between the groups. effect of benfotiamine makes it effective for DN
The ALA studies’ overall results indicate that treatment [143].
chronic treatment with ALA at an optimal dose of Benfotiamine is able to reduce the accumula-
at least 600 mg/day is safe and improves some tion of triosephosphates in diabetes. Excess trios-
neuropathic deficits in patients with DN. However, ephosphates can be removed via the reductive
limitations in end point measures have precluded pentosephosphate pathway. However, this path-
a definitive conclusion about whether ALA way is impaired in diabetes by mild thiamine
reverses or prevents DPN. In the analysis of the deficiency. The activity and expression of the
NATHAN 1 trial, improvement and prevention of thiamine-dependent enzyme, transketolase, are
progression of NIS-LL with ALA vs. placebo consequently decreased in the pentosephosphate
after 4 years was predicted by lower BMI, higher pathway. Benfotiamine therapy in experimental
age, male sex, history of cardiovascular disease diabetes restores disposal of triosephosphates by
(CVD), normal blood pressure, insulin treatment, the reductive pentosephosphate pathway in
longer duration of diabetes and neuropathy, and hyperglycemia. This prevents activation of mul-
Targeting the Mitochondrion in Diabetic Neuropathy 321

tiple biochemical pathways that cause injury in and signaling, although it remains to be seen
diabetes: PKC, hexosamine, glycation and oxida- whether these effects can be sustained. As with
tive stress pathways [144]. High-dose thiamine all interventions, therapeutics targeting mito-
also corrects dyslipidemia in experimental diabe- chondria will face problems with delivery, and
tes, in part by normalizing cholesterol and tri- specificity challenges to ameliorate pathologi-
glycerides. IGT is observed with thiamine cal symptoms without inducing negative side
deficiency, and thus dietary thiamine may help effects.
prevent T2DM.
Benfotiamine has been assessed as a clinical Acknowledgments Supported in part by the National
therapy in a double-blind, placebo-controlled, Institute of Diabetes and Digestive and Kidney Diseases,
National Institutes of Health 1R01DK107007-01A1,
phase-III study [145]. A total of 165 patients with Office of Research Development, Department of Veterans
DPN were randomized to one of three treatment Affairs (Biomedical and Laboratory Research Service and
groups: benfotiamine 600 mg/day, benfotiamine Rehabilitation Research and Development,
300 mg/day, or placebo. After 6 weeks of treat- 101RX001030), Diabetes Action Research and Education
Foundation, University of Maryland Institute for Clinical
ment, the primary outcome parameter, the & Translational Research (ICTR), and the Baltimore
Neuropathy Symptom Score, differed GRECC (JWR), 1K2RX001651 (LAZ).
­significantly between the treatment groups in the
per-­protocol but not in the intention to treat popu-
lation (p = 0.055). The Total Symptom Score References
showed no significant differences after 6 weeks
of treatment. The improvement was more notice- 1. Vincent AM, Olzmann JA, Brownlee M, Sivitz WI,
able at the higher benfotiamine dose and Russell JW.  Uncoupling proteins prevent glucose-­
induced neuronal oxidative stress and programmed
increased with treatment duration. In the TSS, cell death. Diabetes. 2004;53:726–34.
“pain” showed the best response to treatment. 2. Chandrasekaran K, Anjaneyulu M, Choi J, Kumar
Treatment was well tolerated in all groups. P, Salimian M, Ho CY, et al. Role of mitochondria
in diabetic peripheral neuropathy: Influencing the
NAD(+)-dependent SIRT1-PGC-1alpha-TFAM
pathway. Int Rev Neurobiol. 2019;145:177–209.
4 Conclusion 3. Chandrasekaran K, Anjaneyulu M, Inoue T, Choi J,
Sagi AR, Chen C, et al. Mitochondrial transcription
Many lines of evidence indicate that aberrant factor A regulation of mitochondrial degeneration
in experimental diabetic neuropathy. Am J Physiol
mitochondrial structure and function contrib- Endocrinol Metab. 2015;309(2):E132–E41.
ute to cellular dysfunction, cell death, and dis- 4. Choi J, Chandrasekaran K, Inoue T, Muragundla A,
ease pathology including diabetic neuropathy. Russell JW.  PGC-1α regulation of mitochondrial
In general, disease may occur within several degeneration in experimental diabetic neuropathy.
Neurobiol Dis. 2014;64:118–30.
levels of mitochondrial dysfunction. These 5. Rumora AE, Savelieff MG, Sakowski SA, Feldman
include impaired oxidative phosphorylation, EL. Disorders of mitochondrial dynamics in periph-
lipid metabolism, imbalanced mitochondrial eral neuropathy: clues from hereditary neuropathy
dynamics, fragmented mitochondrial networks, and diabetes. Int Rev Neurobiol. 2019;145:127–76.
6. Fernyhough P.  Mitochondrial dysfunction in dia-
impaired mitochondrial signaling, neuroin- betic neuropathy: a series of unfortunate metabolic
flammation, and mutations in the mitochon- events. Curr Diab Rep. 2015;15(11):89–0671.
drial or nuclear genome. Exploration of some 7. Russell JW, Berent-Spillson A, Vincent AM,
of the mitochondrial directed mechanisms for Freimann CL, Sullivan KA, Feldman EL. Oxidative
injury and neuropathy in diabetes and impaired glu-
diabetic neuropathy reveals targets for treat- cose tolerance. Neurobiol Dis. 2008;30(3):420–9.
ment of disease. If these targets are promising 8. Cowell RM, Kennedy E, Berent-Spillson A,
in preclinical studies, they may be utilized in Sundkvist G, Russell JW.  Regulation of mitochon-
human clinical trials. Dietary and exercise drial biogenesis in Schwann cells: implications for
diabetic neuropathy. Ann Neurol. 2005;58:S29.
interventions are not only effective but have 9. Russell JW, Golovoy D, Vincent AM, Mahendru P,
significant effects on mitochondrial function Olzmann JA, Mentzer A, et al. High glucose-induced
322 A. Hedayat et al.

oxidative stress and mitochondrial dysfunction in and other complications in diabetes. Handb Clin
neurons. FASEB. 2002;16(13):1738–48. Neurol. 2014;126:353–77.
10. Vincent AM, Calabek B, Roberts L, Feldman 24. Callaghan BC, Cheng HT, Stables CL, Smith AL,
EL.  Biology of diabetic neuropathy. Handb Clin Feldman EL.  Diabetic neuropathy: clinical mani-
Neurol. 2013;115:591–606. festations and current treatments. Lancet Neurol.
11. Puigserver P. Tissue-specific regulation of metabolic 2012;11(6):521–34.
pathways through the transcriptional coactivator 25. Vincent AM, Callaghan BC, Smith AL, Feldman
PGC1-alpha. Int J Obes. 2005;29(1):5–9. EL.  Diabetic neuropathy: cellular mecha-
12. Puigserver P, Spiegelman BM.  Peroxisome nisms as therapeutic targets. Nat Rev Neurol.
proliferator-­activated receptor-gamma coactivator 1 2011;7(10):573–83.
alpha (PGC-1 alpha): transcriptional coactivator and 26. Vincent AM, Hinder LM, Pop-Busui R, Feldman
metabolic regulator. Endocr Rev. 2003;24(1):78–90. EL.  Hyperlipidemia: a new therapeutic target
13. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, for diabetic neuropathy. J Peripher Nerv Syst.
Jager S, et al. Suppression of reactive oxygen species 2009;14(4):257–67.
and neurodegeneration by the PGC-1 transcriptional 27. Vincent AM, Hayes JM, McLean LL, Vivekanandan-­
coactivators. Cell. 2006;127(2):397–408. Giri A, Pennathur S, Feldman EL.  Dyslipidemia-­
14. Johri A, Calingasan NY, Hennessey TM, Sharma induced neuropathy in mice: the role of oxLDL/
A, Yang L, Wille E, et  al. Pharmacologic activa- LOX-1. Diabetes. 2009;58(10):2376–85.
tion of mitochondrial biogenesis exerts wide- 28. Heilbronn LK, Gregersen S, Shirkhedkar D, Hu D,
spread beneficial effects in a transgenic mouse Campbell LV.  Impaired fat oxidation after a single
model of Huntington’s disease. Hum Mol Genet. high-fat meal in insulin-sensitive nondiabetic indi-
2012;21(5):1124–37. viduals with a family history of type 2 diabetes.
15. Scarpulla RC, Vega RB, Kelly DP.  Transcriptional Diabetes. 2007;56(8):2046–53.
integration of mitochondrial biogenesis. Trends 29. Kleiner S, Mepani RJ, Laznik D, Ye L, Jurczak MJ,
Endocrinol Metab. 2012;23(9):459–66. Jornayvaz FR, et  al. Development of insulin resis-
16. Wu Z, Puigserver P, Andersson U, Zhang C, tance in mice lacking PGC-1alpha in adipose tissues.
Adelmant G, Mootha V, et  al. Mechanisms con- Proc Natl Acad Sci U S A. 2012;109(24):9635–40.
trolling mitochondrial biogenesis and respiration 30. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre
through the thermogenic coactivator PGC-1. Cell. J, Sihag S, et al. Erralpha and Gabpa/b specify PGC-­
1999;98(1):115–24. 1alpha-­dependent oxidative phosphorylation gene
17. Esterbauer H, Oberkofler H, Krempler F, Patsch expression that is altered in diabetic muscle. Proc
W. Human peroxisome proliferator activated recep- Natl Acad Sci U S A. 2004;101(17):6570–5.
tor gamma coactivator 1 (PPARGC1) gene: cDNA 31. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R,
sequence, genomic organization, chromosomal Kashyap S, et al. Coordinated reduction of genes of
localization, and tissue expression. Genomics. oxidative metabolism in humans with insulin resis-
1999;62(1):98–102. tance and diabetes: Potential role of PGC1 and NRF1.
18. Andrulionyte L, Kuulasmaa T, Chiasson JL, Laakso Proc Natl Acad Sci U S A. 2003;100(14):8466–71.
M.  Single nucleotide polymorphisms of the per- 32. Leone TC, Lehman JJ, Finck BN, Schaeffer PJ,
oxisome proliferator-activated receptor-alpha gene Wende AR, Boudina S, et al. PGC-1a deficient mice
(PPARA) influence the conversion from impaired exhibit multi-system energy metabolic derange-
glucose tolerance to type 2 diabetes: the STOP-­ ments: muscle dysfunction, abnormal weight con-
NIDDM trial. Diabetes. 2007;56(4):1181–6. trol, and hepatic steatosis. PLoS Biol. 2005;3:e101.
19. Mootha VK, Lindgren CM, Eriksson KF, 33. Farge G, Mehmedovic M, Baclayon M, van den
Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-­ Wildenberg SM, Roos WH, Gustafsson CM, et  al.
responsive genes involved in oxidative phosphory- In vitro-reconstituted nucleoids can block mitochon-
lation are coordinately downregulated in human drial DNA replication and transcription. Cell Rep.
diabetes. Nat Genet. 2003;34(3):267–73. 2014;8(1):66–74.
20. Roy Chowdhury SK, Smith DR, Saleh A, 34. Alam TI, Kanki T, Muta T, Ukaji K, Abe Y, Nakayama
Schapansky J, Marquez A, Gomes S, et al. Impaired H, et  al. Human mitochondrial DNA is packaged
adenosine monophosphate-activated protein kinase with TFAM. Nucleic Acids Res. 2003;31(6):1640–5.
signalling in dorsal root ganglia neurons is linked to 35. Ohgaki K, Kanki T, Fukuoh A, Kurisaki H, Aoki
mitochondrial dysfunction and peripheral neuropa- Y, Ikeuchi M, et  al. The C-terminal tail of mito-
thy in diabetes. Brain. 2012;135(6):1751–66. chondrial transcription factor a markedly strength-
21. Chowdhury SK, Smith DR, Fernyhough P. The role ens its general binding to DNA.  J Biochem.
of aberrant mitochondrial bioenergetics in diabetic 2007;141(2):201–11.
neuropathy. Neurobiol Dis. 2013;51:56–65. 36. Wang YE, Marinov GK, Wold BJ, Chan
22. Canto C, Auwerx J.  Calorie restriction: is DC.  Genome-wide analysis reveals coating of
AMPK a key sensor and effector? Physiology. the mitochondrial genome by TFAM.  PLoS One.
2011;26(4):214–24. 2013;8(8):e74513.
23. Fernyhough P, Jonathan M. Mechanisms of disease: 37. Morimoto N, Miyazaki K, Kurata T, Ikeda Y,
mitochondrial dysfunction in sensory neuropathy Matsuura T, Kang D, et al. Effect of mitochondrial
Targeting the Mitochondrion in Diabetic Neuropathy 323

transcription factor a overexpression on motor neu- 53. Agca CA, Tuzcu M, Hayirli A, Sahin K.  Taurine
rons in amyotrophic lateral sclerosis model mice. J ameliorates neuropathy via regulating NF-kappaB
Neurosci Res. 2012;90(6):1200–8. and Nrf2/HO-1 signaling cascades in diabetic rats.
38. Murphy MP.  How mitochondria produce reactive Food Chem Toxicol. 2014;71:116–21.
oxygen species. Biochem J. 2009;417(1):1–13. 54. Sandireddy R, Yerra VG, Komirishetti P, Areti A,
39. Chandrasekaran K, Salimian M, Konduru SR, Kumar A. Fisetin imparts neuroprotection in experi-
Choi J, Kumar P, Long A, et  al. Overexpression mental diabetic neuropathy by modulating Nrf2
of Sirtuin 1 protein in neurons prevents and and NF-kappaB pathways. Cell Mol Neurobiol.
reverses experimental diabetic neuropathy. Brain. 2016;36(6):883–92.
2019;142(12):3737–52. 55. Maiese K.  Novel nervous and multi-system
40. Chandrasekaran K, Chen C, Sagi AR, Russell JW. A regenerative therapeutic strategies for diabe-
nicotinamide adenine nucleotide (NAD+) precur- tes mellitus with mTOR.  Neural Regen Res.
sor is a potential therapy for diabetic neuropathy. J 2016;11(3):372–85.
Neuromusc Dis. 2016;3(1):S86. 56. Mazucanti CH, Cabral-Costa JV, Vasconcelos AR,
41. Chandrasekaran K, Najimi N, Kumar P, Russell Andreotti DZ, Scavone C, Kawamoto EM. Longevity
JW.  Nicotinamide riboside is a potential therapy pathways (mTOR, SIRT, Insulin/IGF-1) as key mod-
for diabetic neuropathy. Neurodiab; Bucharest, ulatory targets on aging and neurodegeneration. Curr
Romania. 2016. Top Med Chem. 2015;15(21):2116–38.
42. Trammell SA, Weidemann BJ, Chadda A, Yorek MS, 57. Cowell RM, Russell JW. Nitrosative injury and anti-
Holmes A, Coppey LJ, et al. Nicotinamide riboside oxidant therapy in the management of diabetic neu-
opposes type 2 diabetes and neuropathy in mice. ropathy. J Investig Med. 2004;52(1):33–44.
Sci Rep. 2016;6:26933. https://doi.org/10.1038/ 58. Stavniichuk R, Shevalye H, Lupachyk S, Obrosov
srep26933.:26933. A, Groves JT, Obrosova IG, et  al. Peroxynitrite
43. Feldman EL, Callaghan BC, Pop-Busui R, Zochodne and protein nitration in the pathogenesis of diabetic
DW, Wright DE, Bennett DL, et al. Diabetic neurop- peripheral neuropathy. Diabetes Metab Res Rev.
athy. Nat Rev Dis Primers. 2019;5(1):41. 2014;30(8):669–78.
44. Sena LA, Chandel NS.  Physiological roles of 59. Muller-Stich BP, Billeter AT, Fleming T, Fischer
mitochondrial reactive oxygen species. Mol Cell. L, Buchler MW, Nawroth PP. Nitrosative stress but
2012;48(2):158–67. not glycemic parameters correlate with improved
45. Milenkovic D, Blaza JN, Larsson NG, Hirst J. The neuropathy in nonseverely obese diabetic patients
enigma of the respiratory chain supercomplex. Cell after Roux-Y gastric bypass. Surg Obes Relat Dis.
Metab. 2017;25(4):765–76. 2015;11(4):847–54.
46. Topf U, Suppanz I, Samluk L, Wrobel L, Boser A, 60. Nigro C, Leone A, Fiory F, Prevenzano I, Nicolo A,
Sakowska P, et al. Quantitative proteomics identifies Mirra P, et al. Dicarbonyl stress at the crossroads of
redox switches for global translation modulation by healthy and unhealthy aging. Cell. 2019;8(7):749.
mitochondrially produced reactive oxygen species. 61. Jiating L, Buyun J, Yinchang Z.  Role of metfor-
Nat Commun. 2018;9(1):324. min on osteoblast differentiation in type 2 diabetes.
47. Pfanner N, Warscheid B, Wiedemann Biomed Res Int. 2019;2019:9203934.
N.  Mitochondrial proteins: from biogenesis to 62. Park S, Kang HJ, Jeon JH, Kim MJ, Lee IK. Recent
functional networks. Nat Rev Mol Cell Biol. advances in the pathogenesis of microvascu-
2019;20(5):267–84. lar complications in diabetes. Arch Pharm Res.
48. Cowell RM, Russell JW.  Peripheral neuropathy 2019;42(3):252–62.
and the Schwann cell. In: Kettenmann H, Ransom 63. Toth C, Rong LL, Yang C, Martinez J, Song F, Ramji
BR, editors. Neuroglia. 2nd ed. Oxford: Oxford N, et al. Receptor for advanced glycation end prod-
University Press; 2004. p. 573–85. ucts (RAGEs) and experimental diabetic neuropathy.
49. Vincent AM, Russell JW, Sullivan KA, Backus C, Diabetes. 2008;57(4):1002–17.
Hayes JM, McLean LL, et al. SOD2 protects neurons 64. Goldin A, Beckman JA, Schmidt AM, Creager
from injury in cell culture and animal models of dia- MA. Advanced glycation end products: sparking the
betic neuropathy. Exp Neurol. 2007;208(2):216–27. development of diabetic vascular injury. Circulation.
50. Mendez MM, Folgado J, Tormo C, Artero A, Ascaso 2006;114(6):597–605.
M, Martinez-Hervas S, et al. Altered glutathione sys- 65. Vincent AM, Perrone L, Sullivan KA, Backus C,
tem is associated with the presence of distal sym- Sastry AM, Lastoskie C, et al. Receptor for advanced
metric peripheral polyneuropathy in type 2 diabetic glycation end products activation injures primary
subjects. J Diabetes Complicat. 2015;29(7):923–7. sensory neurons via oxidative stress. Endocrinology.
51. Kumar A, Mittal R. Nrf2: a potential therapeutic tar- 2007;148(2):548–58.
get for diabetic neuropathy. Inflammopharmacology. 66. Kanwar YS, Sun L, Xie P, Liu FY, Chen S.  A
2017;25(4):393–402. glimpse of various pathogenetic mechanisms
52. Jimenez-Osorio AS, Gonzalez-Reyes S, Pedraza-­ of diabetic nephropathy. Annu Rev Pathol.
Chaverri J. Natural Nrf2 activators in diabetes. Clin 2011;6:395–423. https://doi.org/10.1146/annurev.
Chim Acta. 2015;448:182–92. pathol.4.110807.092150.:395-­423.
324 A. Hedayat et al.

67. Haslbeck KM, Schleicher E, Bierhaus A, Nawroth conduction velocity in type 2 diabetes mellitus.
P, Haslbeck M, Neundorfer B, et  al. The AGE/ Diabetes Metab Syndr. 2016;10(1):135–9.
RAGE/NF-(kappa)B pathway may contribute to the 82. Tan CK, Chong HC, Tan EH, Tan NS.  Getting
pathogenesis of polyneuropathy in impaired glucose ‘Smad’ about obesity and diabetes. Nutr Diabetes.
tolerance (IGT). Exp Clin Endocrinol Diabetes. 2012;2:e29.
2005;113(5):288–91. 83. Berent-Spillson A, Robinson A, Golovoy D, Slusher
68. Anjaneyulu M, Berent-Spillson A, Inoue T, Choi J, B, Rojas C, Russell JW. Protection against glucose-­
Cherian K, Russell JW. Transforming growth factor-­ induced neuronal death by NAAG and GCP II
beta induces cellular injury in experimental diabetic inhibition is regulated by mGluR3. J Neurochem.
neuropathy. Exp Neurol. 2008;211(2):469–79. 2004;89:90–9.
69. Toth C, Rong LL, Yang C, Martinez J, Song F, Ramji 84. Choi DW.  Excitotoxic cell-death. J Neurobiol.
N, et al. Receptor for advanced glycation end prod- 1992;23(9):1261–76.
ucts (RAGEs) and experimental diabetic neuropathy. 85. Miller KE, Richards BA, Kriebel RM. Glutamine-,
Diabetes. 2007;57:1002–17. glutamine synthetase-, glutamate dehydrogenase-
70. Ray R, Juranek JK, Rai V.  RAGE axis in neuroin- and pyruvate carboxylase-immunoreactivities in the
flammation, neurodegeneration and its emerging rat dorsal root ganglion and peripheral nerve. Brain
role in the pathogenesis of amyotrophic lateral scle- Res. 2002;945(2):202–11.
rosis. Neurosci Biobehav Rev. 2016;62:48–55. 86. Cangro CB, Sweetnam PM, Wrathall JR, Haser WB,
71. Morgenstern J, Fleming T, Schumacher D, Eckstein Curthoys NP, Neale JH. Localization of elevated glu-
V, Freichel M, Herzig S, et al. Loss of glyoxalase 1 taminase immunoreactivity in small DRG neurons.
induces compensatory mechanism to achieve dicar- Brain Res. 1985;336(1):158–61.
bonyl detoxification in mammalian schwann cells. J 87. Carozzi VA, Canta A, Oggioni N, Ceresa C,
Biol Chem. 2017;292(8):3224–38. Marmiroli P, Konvalinka J, et  al. Expression and
72. Jack MM, Ryals JM, Wright DE. Characterisation of distribution of ‘high affinity’ glutamate trans-
glyoxalase I in a streptozocin-induced mouse model porters GLT1, GLAST, EAAC1 and of GCPII
of diabetes with painful and insensate neuropathy. in the rat peripheral nervous system. J Anat.
Diabetologia. 2011;54(8):2174–82. 2008;213(5):539–46.
73. Alderson NL, Chachich ME, Youssef NN, Beattie 88. Ohara PT, Vit JP, Bhargava A, Romero M,
RJ, Nachtigal M, Thorpe SR, et al. The AGE inhibi- Sundberg C, Charles AC, et  al. Gliopathic pain:
tor pyridoxamine inhibits lipemia and development when satellite glial cells go bad. Neuroscientist.
of renal and vascular disease in Zucker obese rats. 2009;15(5):450–63.
Kidney Int. 2003;63(6):2123–33. 89. Anjaneyulu M, Berent-Spillson A, Russell
74. Archer AE, Von Schulze AT, Geiger PC.  Exercise, JW.  Metabotropic glutamate receptors (mGluRs)
heat shock proteins and insulin resistance. Philos and diabetic neuropathy. Curr Drug Targets.
Trans R Soc Lond Ser B Biol Sci. 2018;373:1738. 2008;9(1):85–93.
75. Mayer MP, Bukau B.  Hsp70 chaperones: cellular 90. Berent Spillson A, Russell JW. Metabotropic gluta-
functions and molecular mechanism. Cell Mol Life mate receptor regulation of neuronal cell death. Exp
Sci. 2005;62(6):670–84. Neurol. 2003;184:S97–S105.
76. Peterson LB, Blagg BS. To fold or not to fold: modu- 91. Berent-Spillson A, Russell JW.  Metabotropic
lation and consequences of Hsp90 inhibition. Future glutamate receptor 3 protects neurons from
Med Chem. 2009;1(2):267–83. glucose-­induced oxidative injury by increasing intra-
77. Henstridge DC, Whitham M, Febbraio cellular glutathione concentration. J Neurochem.
MA.  Chaperoning to the metabolic party: the 2007;101(2):342–54.
emerging therapeutic role of heat-shock pro- 92. Howarth C, Gleeson P, Attwell D.  Updated energy
teins in obesity and type 2 diabetes. Mol Metab. budgets for neural computation in the neocor-
2014;3(8):781–93. tex and cerebellum. J Cereb Blood Flow Metab.
78. Farmer KL, Li C, Dobrowsky RT. Diabetic periph- 2012;32(7):1222–32.
eral neuropathy: should a chaperone accom- 93. Rangaraju V, Calloway N, Ryan TA. Activity-driven
pany our therapeutic approach? Pharmacol Rev. local ATP synthesis is required for synaptic function.
2012;64(4):880–900. Cell. 2014;156(4):825–35.
79. Dobrowsky RT.  Targeting the diabetic chaperome 94. Misgeld T, Schwarz TL.  Mitostasis in neurons:
to improve peripheral neuropathy. Curr Diab Rep. maintaining mitochondria in an extended cellular
2016;16(8):71. architecture. Neuron. 2017;96(3):651–66.
80. Murphy M, Hickey F, Godson C.  IHG-1 amplifies 95. Moehlman AT, Youle RJ. Mitochondrial quality con-
TGF-beta1 signalling and mitochondrial biogenesis trol and restraining innate immunity. Annu Rev Cell
and is increased in diabetic kidney disease. Curr Dev Biol. 2020;36:265–89.
Opin Nephrol Hypertens. 2013;22(1):77–84. 96. Chowdhury SK, Zherebitskaya E, Smith DR,
81. Hussain G, Rizvi SA, Singhal S, Zubair M, Ahmad Akude E, Chattopadhyay S, Jolivalt CG, et  al.
J. Serum levels of TGF-beta1 in patients of diabetic Mitochondrial respiratory chain dysfunction in dor-
peripheral neuropathy and its correlation with nerve sal root ganglia of streptozotocin-induced diabetic
Targeting the Mitochondrion in Diabetic Neuropathy 325

rats and its correction by insulin treatment. Diabetes. 111. Russell JW, Kaminsky AJ. Oxidative injury in dia-
2010;59(4):1082–91. betic neuropathy. In: Opara E, editor. Nutrition and
97. Zuchner S, Mersiyanova IV, Muglia M, Bissar-­ diabetes: pathophysiology and management. Boca
Tadmouri N, Rochelle J, Dadali EL, et al. Mutations Raton: Taylor & Francis; 2005. p. 381–97.
in the mitochondrial GTPase mitofusin 2 cause 112. Kobilo T, Guerrieri D, Zhang Y, Collica SC, Becker
charcot-marie-tooth neuropathy type 2A. Nat Genet. KG, van Praag H. AMPK agonist AICAR improves
2004;36(5):449–51. cognition and motor coordination in young and aged
98. Kijima K, Numakura C, Izumino H, Umetsu K, mice. Learn Mem. 2014;21(2):119–26.
Nezu A, Shiiki T, et al. Mitochondrial GTPase mito- 113. Chandrasekaran K, Choi J, Arvas MI, Salimian
fusin 2 mutation in Charcot-Marie-Tooth neuropathy M, Singh S, Xu S, et  al. Nicotinamide mono-
type 2A. Hum Genet. 2005;116(1-2):23–7. nucleotide administration prevents experimen-
99. Alexander C, Votruba M, Pesch UE, Thiselton tal diabetes-induced cognitive impairment and
DL, Mayer S, Moore A, et  al. OPA1, encoding a loss of hippocampal neurons. Int J Mol Sci.
dynamin-­related GTPase, is mutated in autosomal 2020;21(11):3756.
dominant optic atrophy linked to chromosome 3q28. 114. Hinder LM, O’Brien PD, Hayes JM, Backus C,
Nat Genet. 2000;26(2):211–5. Solway AP, Sims-Robinson C, et al. Dietary rever-
100. Delettre C, Lenaers G, Griffoin JM, Gigarel N, sal of neuropathy in a murine model of prediabe-
Lorenzo C, Belenguer P, et al. Nuclear gene OPA1, tes and metabolic syndrome. Dis Model Mech.
encoding a mitochondrial dynamin-related protein, 2017;10(6):717–25.
is mutated in dominant optic atrophy. Nat Genet. 115. Cooper MA, Menta BW, Perez-Sanchez C, Jack
2000;26(2):207–10. MM, Khan ZW, Ryals JM, et  al. A ketogenic diet
101. Yu-Wai-Man P, Griffiths PG, Gorman GS, Lourenco reduces metabolic syndrome-induced allodynia
CM, Wright AF, Auer-Grumbach M, et  al. Multi-­ and promotes peripheral nerve growth in mice. Exp
system neurological disease is common in patients Neurol. 2018;306:149–57.
with OPA1 mutations. Brain. 2010;133(3):771–86. 116. Coppey L, Davidson E, Shevalye H, Torres ME,
102. Dyall SD, Brown MT, Johnson PJ.  Ancient inva- Yorek MA.  Effect of dietary oils on peripheral
sions: from endosymbionts to organelles. Science. neuropathy-related endpoints in dietary obese rats.
2004;304(5668):253–7. Diabetes Metab Syndr Obes. 2018;11:117–27.
103. Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier 117. Lewis EJH, Perkins BA, Lovblom LE, Bazinet RP,
AK, et  al. Mitochondrial cardiolipin is required Wolever TMS, Bril V. Effect of omega-3 supplemen-
for Nlrp3 inflammasome activation. Immunity. tation on neuropathy in type 1 diabetes: a 12-month
2013;39(2):311–23. pilot trial. Neurology. 2017;88(24):2294–301.
104. West AP, Shadel GS. Mitochondrial DNA in innate 118. Teodoro JS, Gomes AP, Varela AT, Duarte FV, Rolo
immune responses and inflammatory pathology. Nat AP, Palmeira CM. Uncovering the beginning of dia-
Rev Immunol. 2017;17(6):363–75. betes: the cellular redox status and oxidative stress as
105. Starobova H, Monteleone M, Adolphe C, Batoon starting players in hyperglycemic damage. Mol Cell
L, Sandrock CJ, Tay B, et  al. Vincristine-induced Biochem. 2013;376(1-2):103–10.
peripheral neuropathy is driven by canonical 119. Ang L, Jaiswal M, Martin C, Pop-Busui R. Glucose
NLRP3 activation and IL-1beta release. J Exp Med. control and diabetic neuropathy: lessons from
2021;218(5):e20201452. recent large clinical trials. Curr Diab Rep.
106. Savelieff MG, Feldman EL.  Immune-mediated 2014;14(9):528.
vincristine-­induced neuropathy: unlocking thera- 120. UK Prospective Diabetes Study (UKPDS). Intensive
pies. J Exp Med. 2021;218(5):e20210286. blood-glucose control with sulphonylureas or insu-
107. Jia M, Wu C, Gao F, Xiang H, Sun N, Peng P, et al. lin compared with conventional treatment and risk
Activation of NLRP3 inflammasome in peripheral of complications in patients with type 2 diabetes
nerve contributes to paclitaxel-induced neuropathic (UKPDS 33). Lancet. 1998;352(9131):837–53.
pain. Mol Pain. 2017;13:1744806917719804. 121. Duckworth W, Abraira C, Moritz T, Reda D,
108. Kumar A, Negi G, Sharma SS.  Suppression Emanuele N, Reaven PD, et al. Glucose control and
of NF-kappaB and NF-kappaB regulated oxi- vascular complications in veterans with type 2 dia-
dative stress and neuroinflammation by BAY betes. N Engl J Med. 2009;360(2):129–39.
11-7082 (IkappaB phosphorylation inhibitor) in 122. Azad N, Emanuele NV, Abraira C, Henderson WG,
experimental diabetic neuropathy. Biochimie. Colwell J, Levin SR, et al. The effects of intensive
2012;94(5):1158–65. glycemic control on neuropathy in the VA coopera-
109. Zilliox LA, Russell JW. Physical activity and dietary tive study on type II diabetes mellitus (VA CSDM). J
interventions in diabetic neuropathy: a systematic Diabetes Complicat. 1999;13:307–13.
review. Clin Auton Res. 2019;4:443–55. 123. Albers JW, Herman WH, Pop-Busui R, Feldman EL,
110. Smith AG, Russell JW, Feldman EL, Goldstein Martin CL, Cleary PA, et al. Effect of prior intensive
J, Peltier A, Smith S, et  al. Lifestyle interven- insulin treatment during the Diabetes Control and
tion for prediabetic neuropathy. Diabetes Care. Complications Trial (DCCT) on peripheral neuropa-
2006;29:1294–9. thy in type 1 diabetes during the Epidemiology of
326 A. Hedayat et al.

Diabetes Interventions and Complications (EDIC) II). Alpha Lipoic Acid in Diabetic Neuropathy. Free
Study. Diabetes Care. 2010;33(5):1090–6. Radic Res. 1999;31(3):171–9.
124. Knowler WC, Barrett-Connor E, Fowler SE, 136. Ziegler D, Hanefeld M, Ruhnau KJ, Hasche H,
Hamman RF, Lachin JM, Walker EA, et  al. Lobisch M, Schutte K, et al. Treatment of symptom-
Reduction in the incidence of type 2 diabetes with atic diabetic polyneuropathy with the antioxidant
lifestyle intervention or metformin. N Engl J Med. alpha-lipoic acid: a 7-month multicenter random-
2002;346:393–403. ized controlled trial (ALADIN III Study). ALADIN
125. Diabetes Prevention Program Research Group. The III Study Group. Alpha-Lipoic Acid in Diabetic
10-year cost-effectiveness of lifestyle intervention Neuropathy. Diabetes Care. 1999;22:1296–301.
or metformin for diabetes prevention: an intent-to-­ 137. Ziegler D, Ametov A, Barinov A, Dyck PJ, Gurieva
treat analysis of the DPP/DPPOS.  Diabetes Care. I, Low PA, et  al. Oral treatment with alpha-lipoic
2012;35(4):723–30. acid improves symptomatic diabetic polyneu-
126. Singleton JR, Smith AG, Russell JW, Feldman ropathy: the SYDNEY 2 trial. Diabetes Care.
EL.  Microvascular complications of impaired glu- 2006;29(11):2365–70.
cose tolerance. Diabetes. 2003;52:2867–76. 138. Ziegler D, Low PA, Litchy WJ, Boulton AJ, Vinik
127. Balducci S, Iacobellis G, Parisi L, Di Biase N, AI, Freeman R, et al. Efficacy and safety of antioxi-
Calandriello E, Leonetti F, et al. Exercise training can dant treatment with alpha-lipoic acid over 4 years
modify the natural history of diabetic peripheral neu- in diabetic polyneuropathy: the NATHAN 1 trial.
ropathy. J Diabetes Complicat. 2006;20(4):216–23. Diabetes Care. 2011;34(9):2054–60.
128. Singleton JR, Marcus RL, Jackson JE, Lessard K, 139. Ziegler D, Schatz H, Conrad F, Gries FA, Ulrich H,
Graham TE, Smith AG.  Exercise increases cutane- Reichel G. Effects of treatment with the antioxidant
ous nerve density in diabetic patients without neu- alpha-lipoic acid on cardiac autonomic neuropathy
ropathy. Ann Clin Transl Neurol. 2014;1(10):844–9. in NIDDM patients. A 4-month randomized con-
129. Singleton JR, Marcus RL, Lessard MK, Jackson JE, trolled multicenter trial (DEKAN Study). Deutsche
Smith AG.  Supervised exercise improves cutane- Kardiale Autonome Neuropathie. Diabetes Care.
ous reinnervation capacity in metabolic syndrome 1997;20(3):369–73.
patients. Ann Neurol. 2015;77(1):146–53. 140. Ziegler D, Nowak H, Kempler P, Vargha P, Low
130. Kluding PM, Pasnoor M, Singh R, D'Silva LJ, Yoo PA. Treatment of symptomatic diabetic polyneurop-
M, Billinger SA, et al. Safety of aerobic exercise in athy with the antioxidant alpha-lipoic acid: a meta-­
people with diabetic peripheral neuropathy: single-­ analysis. Diabet Med. 2004;21(2):114–21.
group clinical trial. Phys Ther. 2015;95(2):223–34. 141. Didangelos T, Karlafti E, Kotzakioulafi E,
131. Kluding PM, Pasnoor M, Singh R, Jernigan S, Farmer Kontoninas Z, Margaritidis C, Giannoulaki P, et al.
K, Rucker J, et al. The effect of exercise on neuro- Efficacy and safety of the combination of superoxide
pathic symptoms, nerve function, and cutaneous dismutase, alpha lipoic acid, vitamin B12, and carni-
innervation in people with diabetic peripheral neu- tine for 12 months in patients with diabetic neuropa-
ropathy. J Diabetes Complicat. 2012;26(5):424–9. thy. Nutrients. 2020;12(11):3254.
132. Ikuta N, Chikamoto K, Asano Y, Yasui Y, Yokokawa 142. Didangelos T, Karlafti E, Kotzakioulafi E, Margariti
H, Terao K, et al. Time course effect of r-alpha-lipoic E, Giannoulaki P, Batanis G, et  al. Vitamin B12
acid on cellular metabolomics in cultured hepatoma supplementation in diabetic neuropathy: a 1-year,
cells. J Med Food. 2017;20(3):211–22. randomized, double-blind, placebo-controlled trial.
133. Karalis DT, Karalis T, Karalis S, Kleisiari AS, Nutrients. 2021;13(2):395.
Malakoudi F, Maimari KEV.  The effect of alpha-­ 143. Balakumar P, Rohilla A, Krishan P, Solairaj P,
lipoic acid on diabetic peripheral neuropathy and the Thangathirupathi A.  The multifaceted therapeu-
upcoming depressive disorders of type II diabetics. tic potential of benfotiamine. Pharmacol Res.
Cureus. 2021;13(1):e12773. 2010;61(6):482–8.
134. Papanas N, Ziegler D. Efficacy of alpha-lipoic acid 144. Thornalley PJ. The potential role of thiamine (vita-
in diabetic neuropathy. Expert Opin Pharmacother. min B1) in diabetic complications. Curr Diabetes
2014;15(18):2721–31. Rev. 2005;1(3):287–98.
135. Reljanovic M, Reichel G, Rett K, Lobisch M, 145. Stracke H, Gaus W, Achenbach U, Federlin K,
Schuette K, Moller W, et  al. Treatment of diabetic Bretzel RG. Benfotiamine in diabetic polyneuropa-
polyneuropathy with the antioxidant thioctic acid thy (BENDIP): results of a randomised, double
(alpha-lipoic acid): a two year multicenter random- blind, placebo-controlled clinical study. Exp Clin
ized double-blind placebo-controlled trial (ALADIN Endocrinol Diabetes. 2008;116(10):600–5.
Diabetic Sensory Neurons, Dorsal
Root Ganglia, and Neuropathy

Aparna Areti and Douglas W. Zochodne

1 Introduction to injury, risk of falls, intractable neuropathic


pain, and foot ulceration leading to amputation
Worldwide, there are few more compelling clini- [3–10]. From a neurosciences perspective, there
cal problems than burgeoning diabetes mellitus is also exquisite neurobiology, not well under-
(DM) and its irreversible complications, particu- stood, that dictates how this condition, a unique
larly neurological. Diabetic polyneuropathy form of sensory predominant axon neurodegen-
(DPN) is not only the most common DM compli- eration, develops. DPN speaks to the singular
cation, but it is also the most common form of behavior of sensory axon terminals in the epider-
damage to the peripheral nervous system and its mis that possess a dynamic plasticity required to
prevalence dwarfs that of many other neurologi- maintain their investment of skin and its turnover
cal disorders. Over 6% of Canadians have DM, of epidermal keratinocytes [11]. In DPN, this
and the WHO estimates that 366 million people plasticity is abnormal and sensory axons change
worldwide will develop it within 10 years, with their phenotype, retract, and disappear. Human
diabetic polyneuropathy (DPN) in over 50% [1, DPN is described as “stocking and glove”
2]. It is a progressive irreversible neurological because the distal terminals of sensory axons that
problem that begins with foot numbness and innervate the skin of the toes and fingers are
pain. Investigators of this unfortunate disorder retracted first (see our reviews [12–16]). Essential
recognize the urgency of finding new solutions, features of DPN are slowing of conduction veloc-
none successful to date, to prevent DPN’s major ity, altered sensory behavior, and prominent loss
clinical burdens: loss of sensation, insensitivity of dermal and epidermal skin axons [17–20].

A. Areti · D. W. Zochodne (*)


Peripheral Nerve Research Laboratory, Division of
Neurology, Department of Medicine and the
Neurosciences and Mental Health Institute,
University of Alberta, Edmonton, AB, Canada
e-mail: zochodne@ualberta.ca

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 327
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_18
328 A. Areti and D. W. Zochodne

The material in this chapter overlaps with a DRGs is higher than that of the endoneurium of
number of papers and reviews from our labora- peripheral nerve trunks, almost double in a range
tory alone or collaboratively including some of 35–40  mL/100g/min compared to
published recently, which the reader is referred 15–17 mL/100g/min in the endoneurium respec-
to for greater depth [7, 14, 16, 21–27]. These tively. Moreover, there is evidence of partial
related reviews cover similar material but are “autoregulation” of flow, with relative stability
updated in this chapter with several nuances in down to mean arterial pressures of 60  mmHg.
their interpretation. Some of the materials on This differs from the nerve trunk endoneurium
molecular alterations in diabetic neurons are not where a gradual curvilinear relationship between
replicated in this review. In discussing how DM mean arterial pressure and blood flow is observed.
targets the nervous system, we begin with dorsal It is also different than CNS, where flows are
root ganglia, critical but understudied sites that more tightly preserved at low mean arterial blood
house the cell bodies of peripheral sensory pressures. Mean oxygen tensions directly mea-
neurons. sured in DRGs were comparable to the endoneu-
rium but lower than brain. While speculative,
these unique characteristics of DRG physiology
2 Structure and Physiology may render them susceptible to microangiopathy
of Dorsal Root Ganglia in DM.  A second important characteristic of
DRGs is that their barrier characteristics to
Dorsal root ganglia (DRGs) are paired paraspinal ingress of blood-borne molecules are less robust
collections of peripheral neurons, cell bodies than both the blood nerve and blood brain barri-
(perikarya) of sensory axons that supply sensa- ers [31]. This is important translational informa-
tion to the head, trunk, and limbs. Identified tion given that yet undeveloped therapies of DPN
within or adjacent to spinal foramina, they are might be capable of accessing DRG neurons
susceptible to mechanical compression by disc through a “leaky” barrier following systemic
material or osteophytes, an outcome that may be administration.
important in common symptoms such as back or Dorsal root ganglia house populations of
neck discomfort. DRGs have a vascular supply large, medium, and smaller sensory neurons, the
subserved by segmental radicular branches that perikaryal size of which determines the caliber
supply nerve roots and by anastomoses from the of their axons. These neurons are classified as
spinal artery [28]. They are unique structures, not “pseudo-unipolar” because one perikaryal
often studied in humans or animals because of branch forms into a central branch that enters
their relative inaccessibility to dissection. the spinal cord and a peripheral branch joins
Covered by a capsule, within is a subjacent layer with motor and autonomic axons to form mixed
of neurons of varying sizes and molecular spinal nerve roots that blend into peripheral
makeup with their processes directed inward nerve trunks. The central branches of large, neu-
toward the center of the ganglion. The capsule is rofilament rich expressing neurons join the dor-
a derivative of the perineurium of the nerve trunk sal columns of the spinal cord where they travel
and has a slightly different structure and proper- upward to the brainstem. In DM, this anatomy is
ties when it surrounds DRG versus exiting nerve important because detailed MR images of the
root axons [29]. Neuron DRG are also placed at spinal cord have identified dorsal column atro-
varying depths within the ganglia interspersed phy, suggesting degeneration of the central
among the exiting axons. While less studied, it is branches of large afferent sensory fibers [32].
inferred that peripheral trigeminal ganglia share Further resolution in these studies will be
characteristics of paraspinal DRGs. While we are required to determine whether column atrophy
aware of few studies on quantitative measures of relates to increased fiber packing, myelin thin-
blood flow in DRGs, the findings have suggested ning, axon loss, or axonal atrophy. Early images
interesting characteristics [30]. Blood flow of rat of DM DRGs have also been captured by MR
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 329

but have not had the resolution yet to clarify on small peptidergic axons. TrkB is the receptor
their changes beyond gross volume alterations for BDNF and NT4/5, and it is expressed on
[33]. Large caliber neurons give rise to large medium sized neurons, whereas TrkC, the
myelinated “Aα” axons that subserve discrimi- receptor for NT3, is found on larger caliber sen-
native touch, vibration, proprioception, and hair sory neurons. IB4 neurons are responsive to
movement. Their peripheral branches, also GDNF and express its receptor, GFRα1.
myelinated, innervate sensory organs such as
Pacinian corpuscles, Meissner’s corpuscles, and
hair follicles transmitting rapidly adapting 3 DRG Responses to Injury
mechanical sensation. The central branches of
alternative, small neurofilament poor neurons DRG neurons sense injury to their distal axon
synapse within the dorsal horn of the spinal branches by developing a series of structural and
cord. All sensory neurons express neurofila- molecular changes known as the “axotomy”
ments, but the content of this scaffold protein response or axon reaction [41]. Structural
varies with neuronal size, in turn influencing its changes include a change in the histological
signal in studies using immunohistochemical appearance called central “chromatolysis,” attrib-
labeling, hence the inaccurate terminology of uted to displacement of ribosomes to the periph-
“neurofilament negative” small neurons. Small ery of the perikarya. Nuclei similarly are shifted
neurons send unmyelinated axons to the skin to the periphery of the cell. More important are a
and other target organs. In the skin, the axons series of molecular changes initially described by
invade and innervate the epidermis, sensing Richardson, Hokfelt, Verge, and others [40, 42,
pain, mechanical damage, and probably chemo- 43] known as “RAGs” or regeneration-associated
sensitivity. The detailed physiology and inner- genes. Prominent and early characterized RAGs
vation of sensory neurons in the skin are were downregulation of neurofilament proteins,
reviewed elsewhere [34, 35]. Unmyelinated Trk receptors and upregulation of tubulin,
afferent sensory axons, so-­called C fibers that GAP43,c-Jun, and others. More recent RNA
innervate the epidermis as free nerve endings, sequencing data has identified an extensive list of
are divided into two categories, peptidergic and upregulated and downregulated sensory neuron
nonpeptidergic. Peptidergic axons and neurons transcripts [44]. In turn, the trigger for RAG
express substance P (SP) and CGRP (calcitonin development is likely a retrograde signal from
gene-related peptide) and ­terminate in deeper the injured axon, initially an electrical and cal-
layers of the skin known as the stratum basale. cium wave, then retrogradely transported signals
Nonpeptidergic axons are also called Mrgprs including importins α and β and pErk [45]. The
(Mas-related g protein-coupled receptors) or major conceptual idea behind altered gene regu-
IB4 (named for lectin binding that labels these lation after axotomy is that it represents a major
neurons) [36]. This is of interest because in DM, shift in sensory axons from a stable maintenance
peptidergic and nonpeptidergic central axons role to a growth and regenerative state. Both
entering the dorsal horn of the spinal cord may upregulation and downregulation of neuron
be targeted differentially [37]. Medium caliber mRNAs may be important in facilitating a growth
neurons contain CGRP and through intermedi- response. It is also a characteristic of the CNS
ate caliber myelinated axons (Aβ and γ) trans- that similar retrograde RAG induction is attenu-
mit mechanical sensitivity and pain. While the ated, in concert with a dramatically diminished
normal differential protein content of DRG sen- regenerative potential.
sory neurons has expanded, not reviewed here, An important reason for addressing RAGs in
other common classifications include their this review is the remarkable absence of RAG
expression of neurotrophin family receptors development in experimental diabetes despite an
[38–40]. TrkA (Tropomyosin receptor kinase “axotomy-like” loss of their distal terminals.
A), the high affinity NGF receptor, is expressed Missing in particular are the upregulation of
330 A. Areti and D. W. Zochodne

growth-related proteins such as tubulin and cultures without a connection to the systemic
GAP43 [19]. The general downturn in gene circulation. Macrophages are important sources
expression has suggested a degenerative pheno- of growth factors, of relevance if their endoge-
type and the possibility that mRNA generation nous function within DRGs were impaired by
and protein translation are impaired. DM.  Schwann cell (SC) dysfunction, not dis-
There are other remarkable features of DRGs, cussed in this review, is a well-established fea-
but of unknown significance in DM, given the ture of DPN, making it highly likely that related
paucity of studies of both human and chronic perineuronal satellite cells are similarly targeted
animal models. DRG neurons are intimately [47–49].
surrounded by a special class of glial cell known The Sox2 expressing cell populations in
as the perineuronal satellite cells. These are DRG likely overlap with progenitor cells in
placed in close circumferential relationship to spheroids isolated from DRGs [50] and are
DRG sensory neurons and have the curious interesting in that they may offer avenues to
property of enlarging and proliferating in replenish DRG sensory neurons when lost.
response to distal axotomy. This is despite the However at present, there is insufficient evi-
lack of a direct cellular connection to the injured dence that this would be required in
neuron. These glial cells thereby “sense” distal DPN.  Overall, the extent and potential role of
neuronal injury albeit by unclear mechanisms. altered DRG cellular dynamics and signaling
Despite the lack of an overt physical connec- have not been explored in DM.
tion, perineuronal satellite cells have direct One important outcome of DM may be DNA
molecular communication with their neuron damage, either nuclear or mitochondrial. BRCA1
partners. For example, administration of a fluo- (Breast Cancer 1) is a DNA repair molecule that
rochrome dye to distal axon branches not only is repairs double-strand DNA breaks but also has
retrogradely transported to their parent neuron transcriptional activity, acts as a ubiquitin ligase,
perikarya, but over time, the perineuronal glial and has heterochromatin-related gene silencing
cells also express the fluorochrome, having been activities [51, 52]. Surprising new findings indi-
transferred from neurons. Perineuronal satellite cate that axotomy alone is sufficient to trigger a
cells are more dynamic than their neuron part- DNA damage response in DRG sensory neurons
ners and exhibit ongoing turnover-cell death and and that its repair is important for viable regrowth.
proliferation, even in the absence of injury. They Along these lines, axotomized DRG sensory neu-
also have close relationships with intrinsic resi- rons had a substantial rise in γH2A.X (H2A his-
dent macrophages within DRGs. The latter are a tone family member X; >60% of neurons)
newly recognized constituent of DRGs, broadly expression, a marker of DNA damage, a change
classified as among DRCCs (dorsal root ganglia that indicates a dramatic rise in the extent of
resident cycling cells) [46]. DRCCs are popula- DNA damage after injury. Whether DNA damage
tions of self-­renewing cells that include resident can account for alterations in gene expression in
macrophages as well as satellite glial cells that DPN is speculative and has not been examined.
colabel with Sox2, indicating cells with stem- Nuclear BRCA1 colocalizes in neurons with
like characteristics. Moreover, the close physi- γHSA.X expression where it may offer a repair
cal relationships of these cells with adult DRG and stabilizing action that allows neurons to
neurons suggest an ongoing role in the spatial recover and regenerate. For example, elimination
and temporal arrangements of the of BRCA1 in knockdown experiments impaired
DRG.  Interestingly, despite prior assumptions neurite growth of adult sensory neurons in vitro
that DRG macrophages are largely blood borne, and peripheral nerve regeneration in vivo. Further
the populations that proliferate within DRGs are work is required to discover whether DNA dam-
local. For example, their ongoing replication age may be important in DM, whether it is prop-
stimulated by local CSF1 (colony-­stimulating erly repaired by BRCA, and whether it could also
factor 1) signaling can be identified in explant account for diabetic mitochondriopathy [53, 54].
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 331

4 Dorsal Root Ganglia glia and nerves may adapt to microvascular


and Diabetes Mellitus abnormalities in DM, maintaining essential
perfusion through angiogenesis perhaps trig-
Beyond DM-related declines in overall DRG gered by hypoxia. Recent work has also sug-
volume by MR analysis [33], neither gross gested the possibility of altered capillary
structural alterations of ganglia nor intra-gan- dynamics in chronic DM, but it is uncertain
glion axonal damage have been described in whether these are sufficient to cause neurode-
diabetic DRGs. In a chronic model of mouse generation [60] (Table 1).
DM type 1, perikaryal area and nuclear area Endothelin-1 (ET-1) is a highly potent vaso-
were reduced [55]. Careful nonbiased counting constrictor peptide that when topically applied
of DRG neurons in a long-term models of diabe- over the epineurial vascular plexus induced tem-
tes in rats has not identified evidence of sensory porary ischemic conduction block in nondiabetic
neuron loss [19]: absence of loss in rats of nerves. In this model of ischemia, intense vaso-
12 months diabetes duration and a maximum of constriction of the epineurial circulation impaired
10% loss in mice of 9 months diabetes duration downstream blood flow within the endoneurial
[18]. This finding has argued against reports of compartment. In models of DM however, the
short-term DRG neuron apoptosis in cell culture impact had more significant consequences [62–
models, a finding not substantiated and poten- 64]. ET-1 generated endoneurial infarction and
tially linked to high and nonphysiological acute axonal degeneration in diabetics but not control
glucose exposure [56]. Relative preservation of nondiabetic rats. DM microvascular dysfunction
neuron numbers and axon counts during these in arterial and arteriolar preparations has been
long-term chronic models indicate that neuron linked to potentiated microvascular vasoconstric-
drop out is unlikely to contribute to the tion, in turn attributed to attenuated vasodilata-
phenotype. tion [65, 66]. These physiological alterations
Despite the absence of significant neuron develop in the absence of overt structural micro-
dropout, an important and positive finding, angiopathy but nonetheless may render ischemic
DRGs and sensory neurons are nonetheless tar- damage to diabetic neurons.
geted by chronic DM.  While our group failed Selective DRG ischemia was studied using
to identify declines in nerve blood flow in an topical ET-1-induced vasoconstriction of capsu-
extensive series of DM models, we did identify lar and radicular ganglia feeding vessels. In
significant declines in DRG blood flow in DM, ET-1 generated intense vasoconstriction of
chronic models of DM in rats. This was noted the feeding vessels of ganglia with large and
in 17- to 23-week diabetic BBW rats, a model prolonged declines in local ganglion blood flow,
of type 2 DM [57] and 16-week diabetic type 1 exceeding the changes observed in nondiabetics
STZ diabetic rats [58]. These changes, interest- [67]. Sensory neuron damage was manifest as
ingly, developed in the absence of declines of loss of neurofilaments, dissolution of neurons,
DRG oxygen tensions. While reductions in nuclear TUNEL labeling (indicating apoptosis),
DRG blood flow might contribute to neuronal “nests of Nageotte,” replacing lost neurons, and
stress, they may also arise from declines in chromatolytic changes such as displacement of
oxygen demand, with lesser oxygen extraction. nuclei to the periphery of neuron cell bodies.
In an anatomical study of vascular architecture Intraganglionic axons and downstream sural
in rats (examined in the absence of fixation and sensory axons developed degeneration. These
harvested under anesthesia prior to euthana- findings provided evidence that DRGs are
sia), numbers of perfused microvessels were indeed sensitive to ischemia secondary to
elevated in the endoneurium but not DRG of diabetic-­related changes in microvascular
diabetics [59]. The findings suggest that gan- physiology.
Table 1  Phenotype and interventions in chronic diabetic models, Zochodne Laboratory
332

DM duration MCV SCV Thermal Mechanical Footpad


Title Model (pretreatment) Treatment slowing slowing sensation sensation axons Citation
GLP-1 SW mice 8 weeks 4 weeks, insulin Y Y Loss Loss ± [148]
Type 1 STZ pellets, exendin
Exendin-4 + + + + −
Low dose insulin − − + + −
High dose insulin − − − − −
dbdb 8 weeks Exendin-4 + − + + −
Low dose insulin − − − − −
High dose insulin − − − − −
Intranasal C57B6/L 8 weeks Intranasal Y Y F gain M loss [134]
Insulin, RAGE−/− mice Insulin for
8 weeks
Male IN insulin + + − −
Female IN insulin + + + −
RAGE−/− + + − +
male
RAGE−/− IN insulin ± + − −
DM, PTEN KD CD-1 mice 10 weeks None Y Y Loss Loss [163]
regeneration
Epigenetic studies CD-1 mice 20 weeks None Y Y Loss Loss Loss [73]
STZ
12–16 weeks IN miRNA + + + + +
3 doses/2 weeks
Let-7i
MiR-341 + + + −
Spliceosome C57B6/L 12 weeks Y Y Loss Loss [55]
mice STZ
Intraplantar − + + −
CWC siRNAx4; 28d
endpoint
ES studies, CD-1 male 4 or 8 weeks None ± Y Loss Loss [158]
regeneration mice STZ
HSP27 overexpression C57BL/6 24 weeks Y Y Loss Elevated Loss [61]
mice STZ
A. Areti and D. W. Zochodne
DM duration MCV SCV Thermal Mechanical Footpad
Title Model (pretreatment) Treatment slowing slowing sensation sensation axons Citation
HSP overexpression − + + + −
Plantar insulin 8–12 weeks Paw insulin No change Loss (CD-1 Loss [132]
only)
C57BL/6J +
STZ
dbdb +
CD-1 STZ − + +
ZDF model ZDF rats 16 weeks None Y Y No loss Loss Loss [208]
Type 2
Intrathecal SD male rats 8 weeks IT insulin or Y Y Loss [116,
STZ IGF-1- 4 weeks 133]
0.1 IU insulin + − +
0.2 IU insulin + +
IGF-1 + + +
Spontaneous SW mice 36 weeks None + ±
regression STZ
Neurofilament−/− B6C3 mice 8 weeks None Y (incl Y (incl
DM Nf−/−) Nf−/−)
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy

Chronic DM SD rat STZ 52 weeks None Y Y [19, 209,


210]
Chronic DM SD male rats 26 weeks None Y Y [17]
STZ
Near nerve insulin SD male rats 4 weeks Near nerve insulin Y Y [131]
STZ
0.1 IU 3×/week for 4 + Different
weeks nerve
BBW model Female 20 weeks Y Y [57]
BBW rats
Indomethacin SD male 16 weeks Indomethacin± ± Y [211]
rats STZ Guanethidine
8 weeks
(continued)
333
Table 1 (continued)
334

DM duration MCV SCV Thermal Mechanical Footpad


Title Model (pretreatment) Treatment slowing slowing sensation sensation axons Citation
Indomethacin + +
Guanethidine − − worsens
Indo+Guaneth − −
Sulindac SD male rats 12 weeks Sulindac onset Y Y [58, 212]
STZ 16 weeks Sulindac later
Insulin 1.0 IU
Sulindac onset − +
16 weeks
Sulindac later ± ±
1 month
Insulin − +
Bolded entries are findings in the models, with specific interventionsm; (unbolded) from each study listed below; Y = yes; ‘Loss’ refers to an impairment in the listed modality.
For interventions, +indicates that the intervention did improve this parameter, – had no improvement. Nf -/- are neurofilament lacking animals
A. Areti and D. W. Zochodne
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 335

5 Sensory Neurons in Diabetes contribute to the disease and might be amenable


to intervention. Peptide loss may explain the
As discussed above, there is very little direct evi- alterations in vascular reflexes that accompany
dence for significant sensory neuron loss or drop- DPN [72], and shifts in sodium channel expres-
out in chronic DM despite their susceptibility to sion may contribute to neuropathic pain, trigger-
acute ischemia and their vulnerability to short-­ ing anion overload and degeneration in diabetic
term exposure to high glucose levels. Neither neurons. Finally, heightened expression of PARP,
scenario is informative about their fate during NFκB, and activated caspase-3 demonstrate neu-
long-term diabetes. Human studies of diabetic rons under duress, despite their capacity to sur-
DRGs are limited. An older report by Greenbaum, vive. PARP is an energy-consuming DNA repair
lacking immunohistochemical or newer histo- molecule. However, in light of all of these
logical stains, studied six patients at postmortem changes, the finding that most sensory neurons in
[68]. Only three patients with clinical diabetic chronic DM survive remains important news.
polyneuropathy had evidence of mild neuron loss Restoring function and growth to surviving and
with nests of Nageotte, axon retraction bulbs, and existing neurons is an easier task than replenish-
vacuolation of sensory neurons. Vacuolation ing them.
however may have been a processing artifact More recent RNA microarray analyses of
[69]. In a later and separate report using EM, mRNAs in experimental diabetes have added
Schmidt identified degenerative changes in new targets and information [73, 74]. While rela-
DRGs from both chronic DM and aging includ- tively few of these assays have been carried out,
ing dystrophic proximal axons that accumulated it is likely the lists will vary with the choice of the
neurofilament [70]. DM model and its sex and the duration of hyper-
A previous review from our laboratory catego- glycemia. Moreover, unexplored factors such as
rized known molecular changes in experimental the gut microbiome of housed study rodents may
DM [71] including loss of structural proteins alter findings. Cheng et al. [73] identified a series
such as neurofilament subunits, tubulin, of up- and downregulated mRNAs in chronic
CGRP,SP,PACAP, and other neuropeptides, type 1 DM mice (streptozotocin [STZ] induced)
sodium channel Nav1.8, neurotrophin receptors, of duration 5 months. Prolonged models are pre-
GAP43 and NFκB among others. Upregulated ferred given that chronic human disease requires
molecules included activated caspase-3, Poly a significant exposure to DM, not addressed by
(ADP-ribose) polymerase (PARP), sodium chan- short duration studies of 1–2 months. In the long
nels Nav1.3,1.6,1.9 and subunit β3, Heat shock duration STZ model, theoretical concerns raised
protein HSP27, MAPK second messengers over STZ toxicity are obviated. The list of altered
(ERKs, JNKs, P38), and the insulin receptor. mRNAs in the Cheng model was accompanied
This original list, now expanded, remains incom- by two additional features: (1) upregulation of
plete but has identified a range of molecular alter- GW bodies, cytoplasmic sites of RNA processing
ations associated with chronic DM. The loss of within DRGs; and (2) a concurrent list of differ-
neurofilament structural protein mRNA in DRGs entially altered miRNAs, the short RNA supra-
paralleled loss of neurofilament investment in regulatory system of epigenetic RNA control
distal axons and their associated atrophy [17]. (discussed further below). Both the findings offer
Despite this loss, mice lacking neurofilaments additional and alternative explanations for the
nonetheless develop conduction slowing and altered molecular architecture of DM sensory
accelerated changes of DPN, indicating that this neuron-altered mRNA handling by GW bodies
protein is probably not part of the pathogenesis of and shifted supraregulatory control by miRNAs.
the disease [20]. The loss of growth-related These mechanisms contributing to altered protein
GAP43 and changes in MAPK signaling mole- synthesis may be additive to other disruptions
cules are important in providing impetus to sub- such as altered DNA repair considered above.
sequent ideas that attenuated growth programs However, the findings have added yet another
336 A. Areti and D. W. Zochodne

neurodegenerative mechanism, that of altered DRGs. Similarly understudied, DUSPs have


spliceosomes, considered below. proven of interest in unexpected ways, albeit not
Among the differentially regulated mRNA yet in the context of DPN [76]. Sensory neurons,
“hits” in chronic diabetic DRGs, only a few both injured and noninjured, express isoforms
members have been examined in depth to date. DUSP1 and 4 in both perikarya and axons. Their
Price and Tomlinson identified evidence for a knockdown (KD) in sensory neurons, unlike
role of Txnip (thioredoxin interacting protein) in CWC22, suppresses neurite outgrowth, suggest-
experimental DM [75], upregulation also con- ing a role by DUSPs in supporting normal axonal
firmed in our work [73]. The significance of this “plasticity” of adult neurons. Along these lines,
finding is that the protein may be a mediator of DUSP KD or inhibition intensified axonal dam-
oxidative stress in sensory neurons but also act as age in the setting of capsaicin neurotoxicity, indi-
a tumor suppressor, potentially limiting growth. cating a protective role by DUSPs for axons.
From our array list, we have also focused on two Moreover, in vivo, DUSP knockdown or inhibi-
lesser understood upregulated proteins, DUSPs tion accelerated axonal degeneration following
(dual specificity phosphatases) and CWC22, a axon transection-loss of axon excitability, neuro-
spliceosome protein. filament dissolution, and morphological features
CWC22, an upregulated protein involved in of axon breakdown. DUSPs are part of the
the spliceosome, has proved interesting and rele- recently and partly untangled process of active
vant to DM and DPN in different ways [55]. axonal degeneration, for which several member
CWC22 is largely expressed in sensory neuron proteins have been identified. A protein known as
nuclei, and its knockdown enhanced neurite out- SARM1 (sterile alpha and TIR motif containing
growth of adult sensory neurons in dissociated 1) appears central to axonal degeneration. Along
cultures. This finding suggested that the protein these lines, accelerated axonal degeneration sec-
was a “brake” on neuron growth and plasticity. In ondary to DUSP inhibition or KD was rescued by
a chronic model of DPN in mice, unilateral hind- concurrent SARM1 KD. In DM, DUSP upregu-
paw dosing of CWC siRNA designed to knock- lation within sensory neurons might provide a
down CWC improved ipsilateral sensory compensatory response to incipient axon dam-
conduction and thermal sensation. The contralat- age. There is also early evidence that SARM1
eral paw treated with scrambled control siRNA KD may protect against DPN [77]. The cascade
retained neuropathic abnormalities, indicating a of axonal degeneration, not reviewed here, likely
pathogenic role for CWC22 upregulation in involves mechanisms shared by many neuropa-
DPN. Aberrant CWC action may disrupt the spli- thies; its unraveling has offered several new tar-
ceosome with which it is associated, a nuclear gets for axonal protection, including SARM1
complex that determines the fate of transcribed [78]. Downstream of SARM1, further proteins
mRNAs. Additional spliceosome abnormalities also participate in axon breakdown including cal-
in chronic DPN mice included upregulated num- pains and caspase-6, the latter identified in early
bers of Cajal bodies, sites of RNA splicing in degenerating skin axon profiles [79]. Overall,
nuclei, altered localization of survival motor neu- there are close connections between the reper-
ron (SMN) protein, and abnormal nuclear foci of toire of proteins also found mainly in perikarya
small nuclear ribonucleoprotein particles that nonetheless have major impacts on down-
(snRNPs), also parts of the spliceosome. stream axon survival. A full understanding of
Dysregulated motor neuron SMN is linked to the how DM might impact the full complex of axonal
development of spinal muscular atrophy. degeneration related proteins requires further
DUSPs (dual specificity phosphatases; MAP work, but may offer opportunities to intervene.
kinase phosphatases, MKPs), proteins that inter- Understanding the roles of supraregulatory
act with the MAPK signaling pathway, are a sep- miRNAs in rendering diabetic neuron dysfunc-
arate group of proteins we discovered in our tion has only just begun. Their role might relate
chronic DM mRNA array to be upregulated in to the problem of fitting together diverse pieces
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 337

of the DPN phenotype into a single molecular critical capacity for sensory axons to maintain
trigger. These pieces include, for example, elec- their investment in endorgans like skin. In keep-
trophysiological slowing of motor and sensory ing with this theme, we next discuss several new
conduction, distal loss of sensory axons in the examples of strategies to enhance neuron growth
skin and other targets, axon, perikaryal atrophy, in the setting of damage or disease.
and the generation of neuropathic pain despite
fiber loss. Moreover, the range of mRNA changes
discussed above is difficult to link together 6 Regrowth Strategies
beyond a generalized down run of cellular for Sensory Neurons
machinery. miRNAs characteristically have
many mRNA targets and could be important in Diabetic neurons targeted by diverse metabolic
wide “down tuning” of neurons or alternatively in impacts from DM on sensory neurons may with-
supporting inappropriate upregulation, as in the stand damage through strategies that support
case for CWC22. Cheng et  al. [73] identified a their overall growth properties. In several
series of differentially regulated miRNAs in the instances, these approaches, largely not yet tested
same 5-month-old type 1 cohort of diabetic mice in humans, have remarkable impacts on experi-
that underwent mRNA analysis described above. mental DPN. Proposed mechanisms of DPN are
These mice had well-characterized phenotypic not reviewed here in depth. A key tenet however
features of DPN. One of the most prominent dif- is “neurotoxic stress,” arising from glucotoxicity,
ferentially expressed miRNAs was mmu-Let7i, increased polyol flux, free radical toxicity, nitrer-
reduced by 39% in diabetic DRGs. mmu-Let 7i gic damage, mitochondrial abnormalities, lipid
has a large range of mRNA targets (>900) that toxicity, and deficits in growth factors [80–102].
include IGF-1 and its receptor among 84 other In many putative therapies for DPN, alleviation
growth pathway mRNAs, contrasted to 21 metab- of the impact of one or more of these mechanisms
olism and diabetes pathway mRNAs. Exogenous has been highlighted. An important example has
application of a mmu-Let 7i mimic had a trophic been the long history of aldose reductase inhibi-
impact on adult sensory neurons, encouraging tors (ARIs) to eliminate polyol flux. While ARIs
both neurite outgrowth and branching. Moreover, have had some impact on human disease, interest
intranasal administration of a mmu-Let 7i mimic in their use has waned and none have been fully
improved thermal and mechanical sensation, accepted therapeutically [103, 104].
motor and sensory conduction velocity and epi-
dermal innervation in a model of chronic type 1 Growth Factors and Insulin  The limited impact
DM.  In the same work, intranasal treatment of of traditional growth factors in experimental and
anti-miR to instead tune down an alternative human DPN has been reviewed elsewhere [15,
upregulated miRNA, mmu-miR-341, improved 23]. In the cases of neurotrophins, lack of enthu-
thermal sensation, and conduction velocities. The siasm has stemmed from negative clinical trials,
overall findings lent support to the idea that uncertain dosing, downregulation of their recep-
miRNA manipulation may influence the DPN tor expression in models, and their capability to
phenotype and to the idea that intranasal nucleo- support only subsets of neurons [105]. Hepatic
tides, may have promise in DPN or other disor- growth factor has recently been trialed, albeit
ders, not unlike that offered by intranasal with limited published pretrial preclinical data as
insulin. to its mechanism, with results suggesting an
Several findings described above emphasize impact on human pain [106].
the role of growth and plasticity molecules that
may have impaired actions in DPN. Although the We have previously made the case for exploit-
term “plasticity” has wide use, here we apply it to ing the neurotrophic properties of insulin. Almost
the capacity of neurons to extend processes and 50  years ago, Frazier et  al. in “Nerve Growth
grow or regenerate. This property may provide Factor and Insulin,” published by Science,
338 A. Areti and D. W. Zochodne

emphasized the evolutionary structural similari- may account for neuronal insulin resistance, it is
ties between NGF and insulin [107]. Unlike the possible that combined use with insulin sensitiz-
neurotrophins, of which NGF is a member, insu- ers may overcome this limitation.
lin receptors (IRs) are widely expressed on Glucagon-like peptide 1 (GLP-1) operates by
peripheral neurons and are upregulated in dia- enhancing systemic insulin sensitivity in type 2
betic models. IRs share downstream transduction DM, but its receptors are also expressed on adult
cascades with other growth factors [108–117] sensory neurons, and upregulated in a model of
and on ligation they undergo autophosphoryla- DM type 2 [148]. Moreover, a GLP-1 agonist,
tion, exhibit tyrosine kinase activity, and utilize exendin-4 has trophic actions on adult sensory
downstream activators known as IRS-1 and 2 neurons in vitro, associated with increased neu-
(IRS=insulin receptor substrate) [118, 119]. rite outgrowth. In the setting of only a mild
There is eventual activation of the p85 subunit of impact on glycemia, subcutaneous exendin-4
PI3K, a central growth signal for neurons [119– given over 8 weeks in mice with DM of 2 months
127]. Insulin is associated with increased neurite duration was examined for its impact on DPN. In
outgrowth from adult neurons studied in  vitro contrast to partial impacts of either low- or high-­
[128]. It also improves nerve regeneration in non- dose insulin, exendin-4 improved motor and sen-
diabetic injury models, a property that may be sory conduction velocity in type 1 DM mice and
appropriate for addressing DPN [129, 117]. reversed thermal and mechanical sensory loss.
Either loss of insulin in type 1 DM or resistance Similar benefits, excepting sensory conduction
to its actions in type 2 DM may have a role in the velocity, were noted in type 2 DM mice. There
development of peripheral neuron dysfunction. were no significant impacts on epidermal inner-
Finally, the neuronal actions of insulin as a vation but the shorter model durations limited
growth factor are separate from its impact on gly- this endpoint. The overall findings paralleled
cemia. The preclinical data suggesting a role for similar results in two other laboratories [149,
insulin signaling in DPN are several: low insulin 150] and provide preclinical support for human
doses (that do not alter blood glucose levels) studies of GLP-1 agonism in DPN. Human trials
applied near nerve, within the skin, or intrathe- focused on GLP-1 or its analogs in DPN have not
cally to access DRGs all improve DPN [130– yet been reported.
133]. Intranasal insulin, a route that accesses
DRG through the CSF, improved experimental Supporting Intrinsic Neuronal Growth
DPN [134, 135]. Local corneal insulin restored Properties  That sensory neurons require an
innervation as assessed using corneal confocal ongoing level of “plasticity” or growth in the
microscopy [136]. Insulin-like Growth Factors absence of injury is supported by newer findings.
(IGFs), family members, use similar downstream For example, changes in epidermal and hair fol-
signaling pathways [137–139]. licle innervation may readjust at short notice
While the range of impacts of insulin in pre- from noninvasive stimuli, such as hair shaving
clinical work is impressive, there are potential [11]. In DM models, short-term local hindpaw
barriers to its translational success. Insulin resis- injections of low-dose insulin, signaling dermal
tance is a central deficit in most type 2 DM axon insulin receptors, can rapidly restore axon
patients characterized by normal or high systemic numbers to nondiabetic values [132]. Finally,
insulin levels in relationship to failed insulin-­ human skin biopsies of normal subjects display
mediated glucose uptake by muscle and adipose markers of growth and plasticity on epidermal
tissue [140–143]. We described evidence for axons, indicating evidence of ongoing growth.
insulin “resistance” in neurons, referring to a These included GAP43, Shh, SCG10, and others
deficit in its ability to display trophic actions fol- [151]. Overall, it may be that sensory axons
lowing high dose or prolonged insulin exposure require ongoing growth and plasticity simply to
[144, 145]. These findings were confirmed by deal with the wear and tear of skin and routine
others [146, 147]. Whereas several mechanisms shedding of superficial skin keratinocytes.
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 339

Removing intrinsic “brakes” on neuron lation on sensory axon regrowth [155–157].


growth as a novel strategy to enhance plasticity Additional findings were that ES indeed evoked a
allows consideration of several targets. The idea calcium transient, as occurs during precondition-
has been to unleash the growth potential of stable ing, and that it increased induction of specific
and regeneration reluctant adult sensory neurons RAG mRNAs including GAP43 and tubulin
temporarily and locally. Several “brakes” are while reversing the changes in ATF3, an injury
“tumor suppressor” molecules and pathways marker [158]. In vitro ES enhanced the transcrip-
identified as being gatekeepers to uncontrolled tion of BDNF mRNA, indicating a close connec-
tumor cell growth. Moreover, some “brakes” are tion between ES and preconditioning [156, 159].
not among injury-related “RAGs” described It also lowered PTEN levels (see below) and was
above, indicating that their expression is ongo- sensitive to PI3K inhibition.
ing, and stable irrespective of injury. Others how- Superimposed axon injury in the setting of
ever are tuned down after injury to foster DPN offers insights into the regenerative poten-
regrowth. “Preconditioning” refers to a height- tial of neurons, not unlike those gained by pre-
ened regenerative response of peripheral neurons conditioning. In the peripheral nervous system,
that have been subjected to an axon injury (axot- DM is associated with a “double hit” comprised
omy) within the previous 5–7 days; it ramps up of degenerative neuropathy as well as a specific
early RAG expression, allowing a more robust failure in regeneration. This topic has been
response to a subsequent injury [152]. While the reviewed in detail separately [160]. Nonetheless,
mechanism of preconditioning is unconfirmed, ES, as a surrogate for preconditioning, was asso-
rises in BDNF neurotrophic signaling have been ciated with improved regeneration from a super-
considered. In vitro preparations of adult neurons imposed sciatic nerve injury in chronic diabetic
(see below) demonstrate a dramatic rise in out- mice [158]. Yet to be tested is whether ES reverses
growth if their axons are subjected to an axotomy overall features of DPN, but since this is a focal
prior to their harvesting and growth in culture. strategy, thought will be required as how best to
How dissociated cultured adult neurons respond exploit it for generalized nerve dysfunction in
to interventions in vitro is an important investiga- DM.
tive screening step as a robust predictor of in vivo PTEN (phosphatase and tensin homolog
regeneration. “Neurites” are axon-like out- deleted on chromosome 10) was among the first
growths from neurons in  vitro and their growth tumor suppressor molecules examined in periph-
along with overall neuron survival, neurite initia- eral neurons [161]. Mutations in PTEN are asso-
tion, branching, and directional conduct are all ciated with Cowden’s syndrome, a condition that
important deliverables from in vitro analysis. predisposes persons to oncogenesis with solid
Replicating the molecular reprogramming tumors. PTEN is expressed in adult DRG neu-
generated by preconditioning might encourage rons, but more prominently in small caliber non-
better regrowth of peripheral sensory neurons. peptidergic IB4 or Mpgprs neurons that have
While the triggers for preconditioning are slower regrowth properties [162]. We noted that
debated, axon injuries generate a retrograde PTEN pharmacological inhibition or knockdown
directed injury discharge and a calcium wave. On by siRNA was associated with dramatic rises in
the supposition that exogenous electrical stimula- neurite outgrowth from dissociated adult neurons
tion (ES) might mimic or augment this signal, in vitro. PTEN knockdown or inhibition was an
Gordon, Brushart, and colleagues identified that important example of the impact of removing a
a specific ES paradigm, delivered immediately key “brake” on neuron plasticity, demonstrating
after axotomy to the intact proximal stump was robust enhanced outgrowth of their neurites.
associated with improved motor axon regenera- Moreover, this impact was striking irrespective of
tion and preferential motor branch reinnervation whether the neurons were previously uninjured
of muscles [153, 154]. Follow on work confirmed (intact) or already preconditioned.
a similar benefit from a 1 h epoch of 20 Hz stimu- Preconditioning has been assumed to generate
340 A. Areti and D. W. Zochodne

the peak regenerative response that neurons are in  vitro, in part through increased levels of
capable of. However, both the ES work, superim- PPARϒ [Peroxisome proliferator-activated
posing stimulation on an injury, and PTEN inhi- receptor gamma]. PPARϒ, in turn, was expressed
bition have indicated that it is possible to drive in sensory neuron nuclei and cytoplasm, and it
neuron regenerative plasticity significantly higher enhanced in vitro neurite growth. In addition to
than achieved with preconditioning alone. being a mediator of Rb1 knockdown’s impact on
Indeed, in the case of PTEN inhibition or knock- neurons, PPARϒ is a known insulin sensitizing
down, heightened neurite outgrowth was yet agent [172, 173], recognized to promote insulin
more pronounced if superimposed on an injury signaling at several levels: IRS-1 and IRS-2 [174,
state. Thus, this was the first example we were 175], the p85 subunit of PI3K [176] and else-
aware of in which neuron outgrowth could be where [177, 178]. Given this series of findings,
ramped up substantially by a single molecular Rb1 knockdown, E2F1 activation, and PPARγ
trigger beyond the preconditioning level. In vivo may allow neurons to overcome insulin resis-
PTEN knockdown or inhibition increased axon tance and respond to insulin especially in more
outgrowth from transected sciatic nerves. In both common type 2 DM. This role has yet to be con-
type 1 and type 2 DM mice, PTEN was upregu- firmed. However, preliminary evidence indicates
lated in DRG sensory neurons and its knockdown that Rb1 knockdown has synergistic impacts on
enhanced their neurite outgrowth in vitro. Finally, neuronal growth with insulin and that it improves
we confirmed that PTEN knockdown in  vivo chronic experimental DPN (unpublished data).
could repair the regenerative deficit of mice with There are additional, less well-explored mol-
type 1 DM and others confirmed similar findings ecules and pathways that may offer roles in
in a type 2 DM model in vivo [163, 164]. Taken restoring neuron plasticity. Adenomatous polyp-
together these findings indicate that diabetic neu- osis coli (APC) is mutated in colorectal carcino-
rons are responsive to strategies that improve mas [179] and suppresses tumorigenesis by
their plasticity, an important property to exploit forming a destruction protein complex with
in treatment. β-catenin, also a divergent transcription factor.
The next “tumor suppressor” analyzed was APC thereby blocks β-catenin signaling to sup-
chosen for its central role in transcriptional regu- press growth. APC expression rises in DRG neu-
lation, Rb1(retinoblastoma 1), known to be rons after axotomy injury, and it is expressed in
mutated in childhood retinoblastoma tumors. the sensory neuron cytoplasm, axons, and SCs.
Rb1 is expressed in the cytoplasm and nuclei of Like PTEN, APC is prominent within nonpepti-
adult DRG primary sensory neurons [165], but dergic IB4 neurons that have attenuated growth
unlike PTEN, its expression appears to involve behavior. APC knockdown facilitated β-catenin
most sensory neuron subtypes, including those translocation to the neuron nucleus and enhanced
from mice with chronic DM. In its nonphosphor- neurite outgrowth in vitro and nerve regeneration
ylated state Rb1 binds to a divergent transcription in  vivo. The analysis of its impact on nerve
factor E2F1 and prevents its activation [166]. regrowth involved local application of siRNA at a
While E2F1 signaling is best established in facili- nerve injury site and ipsilateral knockdown of its
tating S phase mitotic DNA replication stage mRNA in the DRG and sciatic nerve. As in sev-
entry, its actions are less well understood in eral siRNA studies to date, the findings speak to
growth [167, 168]. Following phosphorylation by the mobility of small nucleotides exploiting ret-
cyclin-CDKs Rb1 releases E2F1 to permit its sig- rograde transport to knockdown and obviating
naling. Like PTEN, Rb1 is not among the known the need for viral vector directed therapies [180].
RAGs indicating a constitutive “braking” role Sonic hedgehog protein (Shh) is a morphogen
independent of injury or axotomy [44, 165, 169]. that directs developmental motor pathway forma-
Rb1 knockdown also does not activate PI3k-pAkt tion. It is expressed in adult DRG sensory neu-
[161, 170, 171], but its knockdown enhanced rons, axons, and Schwann cells. Unlike the tumor
adult neuron outgrowth, branching and initiation suppressor molecules, constitutive Shh may act
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 341

to support regeneration, as its knockdown attenu- betic mice, the M1R antagonist, pirenzepine
ates growth. Shh has been identified within epi- applied at 2% to the paw improved tactile allo-
dermal axons from normal human subjects, dynia, thermal sensory loss, and loss of epider-
supporting ideas that these axons are normally in mal axons. Interestingly pirenzepine does not
a state of ongoing growth [151]. Finally, but not cross the blood brain barrier and has already been
discussed here are a range of SC mitogens and used extensively as a therapy in the clinic for
signals that may support axon regrowth indirectly reflux and other clinical conditions. A clinical
through the close partnership of axons and SCs trial of pirenzepine topical in patients with type 2
during regrowth [181, 182]. DM is underway.

Topical Manipulation of Sensory Axons  While


this review has focused on overall strategies to 7 Sensory Neurons and Pain
improve the growth and plasticity of sensory neu-
rons, manipulating their distal terminals may be This topic is reviewed in detail elsewhere [15,
of interest. For example, a substantial number of 191, 192]. The pain phenotype of DPN is of com-
mRNAs are transported from perikarya (cell bod- plex origin involving alterations within DRGs,
ies) distally to axon terminals and their growth axons, and their ion channels together with
cones where they undergo local translation [183, altered pain pathway signaling. Within DRG neu-
184]. CGRP is an interesting example of a local rons, the generation of pacemaker ectopic dis-
axon synthesized peptide that signals to SCs and charges, reflecting inappropriate excitability has
microvessels [185, 186]. Several growth factor been recognized for several decades as contribut-
receptors, including the insulin receptor, are also ing to pain [193]. Both peripherally generated
expressed on skin axons and discussed above. and perikaryal originating discharges may under-
Local intracutaneous insulin restored epidermal lie the positive sensory symptoms that contribute
innervation in type 1 and 2 DM models of mice to painful DPN.  Current ideas have particularly
[132]. Beyond these findings, growth cones also focused on the roles of sodium channels and
express a number of intrinsic proteins, not T-type calcium channels. Mutated SCN9A
reviewed here, that determine whether they Nav1.7 sodium channel α subunits that are associ-
advance and grow or collapse and retract. An ated with a gain of function and hyperexcitability
important example is RhoA, a GTPase that acti- have been identified by Faber, Merkies, and
vates ROCK (Rho kinase) to collapse growth Waxman in painful idiopathic sensory polyneu-
cones exposed to inhibitor cues such as myelin or ropathies [194]. Nav1.7 channels are localized to
chondroitin sulfate proteoglycans. RhoA or DRG neurons and axon terminals [195].
ROCK inhibition increases growth of adult sen- Alternative sodium channel α subunits desig-
sory neurons in  vitro and outgrowth of axons nated Nav1.8 are expressed on DRG cell bodies
from a nerve injury [187]. and terminals and have activation properties
linked to Nav1.7. Both have been described in
A novel and exciting approach to directly sup- diabetic models linked to methylglyoxal, a meta-
port regrowth of skin axons has emerged from bolic byproduct of DM [195, 196]. β-Subunits of
work by Fernyhough and Calcutt exploiting axo- the sodium channel that modulate channel open-
nal muscarinic acetylcholine type 1 receptors ing have also been implicated [197] and a spe-
(M1Rs) [188–190]. Mice lacking the receptors cific mutation, associated with neuron
had enhanced neurite outgrowth in vitro and were hyperexcitability, has recently been described in
protected from DPN.  Moreover, M1R-specific a diabetic patient [198].
antagonists reversed mitochondrial dysfunction Similarly, hyperexcitability of DRG neurons
in diabetes and features of neuropathy by mecha- may develop from T-type voltage-gated calcium
nisms operating in part through activation of channels (Cav3.2) that are found in small- and
CAMKKβ, AMPK, and PGC-1α. In female dia- medium-sized neurons. These channels may also
342 A. Areti and D. W. Zochodne

contribute toward ectopic painful discharges in ognition that DPN is a unique neurodegenerative
DM [199, 200]. Their upregulated currents have disorder with sensory, but also autonomic and later
been described in both types 1 and 2 DM models motor neuron targeting. Despite an onslaught of
[201, 202]. multiple mechanisms that generate neurotoxic
Abnormalities in other channels have also “stress” with efforts to mitigate them, this review
been described in DPN including HCN instead emphasizes the promise of enhancing the
(hyperpolarization-­ activated cyclin nucleotide-­ overall plasticity of the sensory neuron, enabling it
gated) channels [203] and potassium ion chan- to withstand diabetes and thrive.
nels, specifically Kv1.2 [204]. Related changes in
the function of TRP channels including TRPV1 Acknowledgments  The effort devoted to this chapter was
and TRPA1 have also been described in relation supported by current operating grants from the Canadian
Institutes of Health Research (FRN148675 and 168929).
to pain from DPN [205, 206]. The authors acknowledge the experimental work cited
Taken together, the evidence for several forms here and contributions by colleagues and trainees in the
of ion channelopathy in DM that contribute to Zochodne laboratory. Work described has been supported
pain is substantial. It is unclear whether axon since 1989 by the Canadian Institutes of Health Research,
Canadian Diabetes Association, the Alberta Heritage
ectopic discharges, DRG neuron pacemaking, or Foundation for Medical Research, Muscular Dystrophy
remodeling of upstream CNS pain pathways are Association of Canada, University of Alberta Hospital
primarily responsible for DM pain syndromes. It Foundation, Department of Medicine and Division of
is recognized that peripherally originating dis- Neurology, University of Alberta, NIDDK Complications
Consortium, and the Juvenile Diabetes Foundation.
charges are capable of widespread secondary
remodeling of ascending nociceptive pathways.
Disclosures  None.
Additional questions one might ask are how spe-
cific ion channel abnormalities develop in overall
DPN degeneration and what mechanisms gener-
ate these changes: transcriptional, translational,
or epigenetic? Since not all DPN phenotypes References
have pain, it seems difficult to attribute neurode- 1. Wild S, Roglic G, Green A, et al. Global prevalence
generation to pain-associated cation overload of diabetes: estimates for the year 2000 and projec-
within axons or perikarya. In DPN models, some tions for 2030. Diabetes Care. 2004;27:1047–53.
mouse strains routinely develop thermal and 2. Capes S, Anand S.  What is type 2 diabetes? In:
Gerstein HC, Haynes RB, editors. Evidence-­
mechanical sensory loss, even early in their based diabetes care. Hamilton: Bc Decker; 2001.
course, whereas others are characterized by p. 151–63.
hyperalgesia. In patients, there may be an impor- 3. Zochodne DW, Said G.  Recombinant human
tant interaction between genotype and the likeli- nerve growth factor and diabetic polyneuropathy.
Neurology. 1998;51:662–3.
hood to develop the painful subtype of DPN. 4. Apfel SC, Kessler JA, Adornato BT, et  al.
Recombinant human nerve growth factor in the
treatment of diabetic polyneuropathy. NGF Study
8 Conclusions Group. Neurology. 1998;51:695–702.
5. Bril V. Invited review: status of current clinical tri-
als in diabetic polyneuropathy. Can J Neurol Sci.
The interaction between sensory neurons and 2001;28:191–8.
chronic DM remains complex and not neatly cat- 6. Brown MJ, Asbury AK.  Diabetic neuropathy. Ann
egorized. Sensory neurons develop a range of mor- Neurol. 1984;15:2–12.
7. Zochodne DW. Diabetic neuropathies: features and
phological (axon retraction), gene expression and mechanisms. Brain Pathol. 1999;9:369–91.
protein changes that are difficult to link into a uni- 8. Dyck PJ, Kratz KM, Karnes JL, et  al. The preva-
fying mechanism. Only briefly mentioned here are lence by staged severity of various types of dia-
primary targeting of mitochondria, reviewed else- betic neuropathy, retinopathy, and nephropathy in
a population-based cohort: the Rochester Diabetic
where [207] and dysfunction of Schwann cells, the Neuropathy Study. Neurology. 1993;43:817–24.
critical neuron glial partner. Important is the rec-
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 343

9. Mulder DW, Lambert EH, Bastron JA, et  al. The molecular targets. American Academy of Neurology
neuropathies associated with diabetes mellitus. Abstracts P2.010. 2015.
Neurology. 1961;11:275–84. 28. Adams WE. The blood supply of nerves. I. Historical
10. Rundles RW.  Diabetic neuropathy—general review review. J Anat. 1942;76:323–41.
with report of 125 cases. Medicine. 1945;24:111–60. 29. Reinhold AK, Rittner H.  Characteristics of the
11. Cheng C, Guo GF, Martinez JA, et al. Dynamic plas- nerve barrier and the blood dorsal root gan-
ticity of axons within a cutaneous milieu. J Neurosci. glion barrier in health and disease. Exp Neurol.
2010;30:14735–44. 2020;327:113244.
12. Zochodne DW. Diabetes mellitus and the peripheral 30. Zochodne DW, Ho LT. Unique microvascular char-
nervous system: manifestations and mechanisms. acteristics of the dorsal root ganglion in the rat.
Muscle Nerve. 2007;36:144–66. Brain Res. 1991;559:89–93.
13. Anderson D, Zochodne DW.  Current ideas on the 31. Arvidson B. Distribution of protein tracers in periph-
treatment of diabetic neuropathies. Expert Rev eral ganglia. A light and electron microscopic study
Endocrinol Metab. 2016;11(2):187–05. in rodents after various modes of tracer administra-
14. Zochodne DW.  Sensory neurodegeneration in dia- tion. Acta Univ Ups. 1979;344:1–72.
betes: beyond glucotoxicity. Int Rev Neurobiol. 32. Selvarajah D, Wilkinson ID, Emery CJ, et al. Early
2016;127:151–80. involvement of the spinal cord in diabetic peripheral
15. Zochodne DW.  Mechanisms of diabetic neuron neuropathy. Diabetes Care. 2006;29:2664–9.
damage: molecular pathways. Handb Clin Neurol. 33. Jende JME, Kender Z, Rother C, et  al. Diabetic
2014;126:379–99. polyneuropathy is associated with pathomorpho-
16. Zochodne DW.  Diabetes and the plasticity of sen- logical changes in human dorsal root ganglia: a
sory neurons. Neurosci Lett. 2015;596:60–5. study using 3T MR neurography. Front Neurosci.
17. Scott JN, Clark AW, Zochodne DW. Neurofilament 2020;14:570744.
and tubulin gene expression in progressive experi- 34. Willis WD, Coggeshall RE.  Sensory mechanisms
mental diabetes: failure of synthesis and export by of the spinal cord. 3rd ed. New  York: Kluwer
sensory neurons. Brain. 1999;122:2109–18. Academic/Plenum; 2004.
18. Kennedy JM, Zochodne DW. Experimental diabetic 35. Zochodne DW. Our wired nerves. The human nerve
neuropathy and spontaneous recovery: is there irrep- connectome. San Diego: Academic Press; 2020.
arable damage? Diabetes. 2005;54:830–7. 36. Zylka MJ, Rice FL, Anderson DJ. Topographically
19. Zochodne DW, Verge VMK, Cheng C, et  al. Does distinct epidermal nociceptive circuits revealed
diabetes target ganglion neurons? Progressive sen- by axonal tracers targeted to Mrgprd. Neuron.
sory neuron involvement in long term experimental 2005;45:17–25.
diabetes. Brain. 2001;124:2319–34. 37. Akkina SK, Patterson CL, Wright DE. Gdnf rescues
20. Zochodne DW, Sun H-S, Cheng C, et al. Accelerated nonpeptidergic unmyelinated primary afferents in
diabetic neuropathy in axons without neurofila- streptozotocin-treated diabetic mice. Exp Neurol.
ments. Brain. 2004;127:2193–200. 2001;167:173–82.
21. Feldman EL, Callaghan BC, Pop-Busui R, et  al. 38. Richardson PM, Verge VMK, Riopelle
Diabetic neuropathy. Nat Rev Dis Primers. RJ.  Quantitative radioautography for NGF recep-
2019;5:41. tors. In: Rush RA, editor. Nerve growth factors.
22. Kobayashi M, Zochodne DW. Diabetic polyneurop- Chichester: Wiley; 1989. p. 315–26.
athy: bridging the translational gap. J Peripher Nerv 39. Verge VMK, Riopelle RJ, Richardson PM.  Nerve
Syst. 2020;25:66–75. growth factor receptors on normal and injured sen-
23. Kobayashi M, Zochodne DW.  Diabetic neuropathy sory neurons. J Neurosci. 1989;9:914–22.
and the sensory neuron: new aspects of pathogenesis 40. Funakoshi H, Frisen J, Barbany G, et al. Differential
and their treatment implications. J Diabetes Investig. expression of mRNAs for neurotrophins and their
2018;9:1239–54. receptors after axotomy of the sciatic nerve. J Cell
24. Zochodne DW.  The challenges of diabetic poly- Biol. 1993;123:455–65.
neuropathy: a brief update. Curr Opin Neurol. 41. Lieberman AR.  The axon reaction: a review of the
2019;32:666–75. principal features of perikaryal responses to axon
25. Zochodne DW.  Is early diabetic neuropathy a injury. Int Rev Neurobiol. 1971;14:49–124.
disorder of the dorsal root ganglion? A hypoth- 42. Richardson PM, Verge VM.  The induction of a
esis and critique of some current ideas on the eti- regenerative propensity in sensory neurons fol-
ology of diabetic neuropathy. J Peripher Nerv Syst. lowing peripheral axonal injury. J Neurocytol.
1996;1:119–30. 1986;15:585–94.
26. Zochodne DW.  Nerve and ganglion blood flow in 43. Verge VMK, Gratto KA, Karchewski LA, et  al.
diabetes: an appraisal. In: Tomlinson D, editor. Neurotrophins and nerve injury in the adult. Philos
Neurobiology of diabetic neuropathy, vol. 50. San Trans R Soc Lond B Biol Sci. 1996;351:423–30.
Diego: Academic Press; 2002. p. 161–202. 44. Chandran V, Coppola G, Nawabi H, et al. A systems-­
27. Kobayashi, M, Cheng, C, De La Hoz, C, et  al. level analysis of the peripheral nerve intrinsic axonal
Diabetic neuropathy and the sensory neuron: new growth program. Neuron. 2016;89:956–70.
344 A. Areti and D. W. Zochodne

45. Perlson E, Medzihradszky KF, Darula Z, et  al. 60. Ostergaard L, Finnerup NB, Terkelsen A, et al. The
Differential proteomics reveals multiple components effects of capillary dysfunction on oxygen and glu-
in retrogradely transported axoplasm after nerve cose extraction in diabetic neuropathy. Diabetologia.
injury. Mol Cell Proteomics. 2004;3:510–20. 2015;58:666–77.
46. Krishnan A, Bhavanam S, Zochodne D.  An inti- 61. Korngut L, Ma CH, Martinez JA, et al.Overexpression
mate role for adult dorsal root ganglia resident of human HSP27 protects sensory neurons from dia-
cycling cells in the generation of local macrophages betes. Neurobiol Dis. 2012;47(3):436–43.
and satellite glial cells. J Neuropathol Exp Neurol. 62. Zochodne DW, Ho LT, Gross PM. Acute endoneu-
2018;77:929–41. rial ischemia induced by epineurial endothelin in the
47. Kalichman MW, Powell HC, Mizisin AP. Reactive, rat sciatic nerve. Am J Physiol. 1992;263:H1806–10.
degenerative, and proliferative Schwann cell 63. Zochodne DW, Cheng C, Sun H. Diabetes increases
responses in experimental galactose and human dia- sciatic nerve susceptibility to endothelin-induced
betic neuropathy. Acta Neuropathol. 1998;95:47–56. ischemia. Diabetes. 1996;45:627–32.
48. Goncalves NP, Vaegter CB, Pallesen LT. Peripheral 64. Zochodne DW, Cheng C. Diabetic peripheral nerves
glial cells in the development of diabetic neuropathy. are susceptible to multifocal ischemic damage from
Front Neurol. 2018;9:268. endothelin. Brain Res. 1999;838:11–7.
49. Gumy LF, Bampton ET, Tolkovsky 65. Zochodne DW, Ho LT.  Diabetes mellitus prevents
AM.  Hyperglycaemia inhibits Schwann cell pro- capsaicin from inducing hyperaemia in the rat sciatic
liferation and migration and restricts regeneration nerve. Diabetologia. 1993;36:493–6.
of axons and Schwann cells from adult murine 66. Thomsen K, Rubin I, Lauritzen M.  NO- and non-
DRG. Mol Cell Neurosci. 2008;37:298–311. NO-, non-prostanoid-dependent vasodilatation in
­
50. Geuna S, Borrione P, Fornaro M, et al. Neurogenesis rat sciatic nerve during maturation and develop-
and stem cells in adult mammalian dorsal root gan- ing experimental diabetic neuropathy. J Physiol.
glia. Anat Rec. 2000;261:139–40. 2002;543:977–93.
51. Starita LM, Parvin JD.  The multiple nuclear func- 67. Xu Q-G, Cheng C, Sun H, et al. Local sensory gan-
tions of BRCA1: transcription, ubiquitination and glion ischemia induced by endothelin vasoconstric-
DNA repair. Curr Opin Cell Biol. 2003;15:345–50. tion. Neuroscience. 2003;122:897–905.
52. Krishnan A, Purdy K, Chandrasekhar A, et  al. A 68. Greenbaum D, Richardson PC, Salmon MV, et  al.
BRCA1-dependent DNA damage response in the Pathological observations on six cases of diabetic
regenerating adult peripheral nerve milieu. Mol neuropathy. Brain. 1964;87:201–14.
Neurobiol. 2018;55(5):4051–67. 69. Li X-G, Zochodne DW.  Microvacuolar neuronopa-
53. Huang TJ, Price SA, Chilton L, et  al. Insulin pre- thy is a post-mortem artifact of sensory neurons. J
vents depolarization of the mitochondrial inner Neurocytol. 2003;32:393–8.
membrane in sensory neurons of type 1 diabetic 70. Schmidt RE, Beaudet LN, Plurad SB, et al. Axonal
rats in the presence of sustained hyperglycemia. cytoskeletal pathology in aged and diabetic
Diabetes. 2003;52:2129–36. human sympathetic autonomic ganglia. Brain Res.
54. Roy CS, Djordjevic J, Thomson E, et al. Depressed 1997;769:375–83.
mitochondrial function and electron transport 71. Toth C, Brussee V, Cheng C, et al. Diabetes mellitus
complex II-mediated H2O2 production in the cor- and the sensory neuron. J Neuropathol Exp Neurol.
tex of type 1 diabetic rodents. Mol Cell Neurosci. 2004;63:561–73.
2018;90:49–59. 72. Caselli A, Rich J, Hanane T, et  al. Role of
55. Kobayashi M, Chandrasekhar A, Cheng C, et  al. C-nociceptive fibers in the nerve axon reflex-­
Diabetic polyneuropathy, sensory neurons, nuclear related vasodilation in diabetes. Neurology.
structure and spliceosome alterations: a role for 2003;60:297–300.
CWC22. Dis Model Mech. 2017;10:215–24. 73. Cheng C, Kobayashi M, Martinez JA, et al. Evidence
56. Russell JW, Sullivan KA, Windebank AJ, et  al. for epigenetic regulation of gene expression and
Neurons undergo apoptosis in animal and cell function in chronic experimental diabetic neuropa-
culture models of diabetes. Neurobiol Dis. thy. J Neuropathol Exp Neurol. 2015;74:804–17.
1999;6:347–63. 74. Price SA, Zeef LA, Wardleworth L, et  al.
57. Zochodne DW, Ho LT, Allison JA.  Dorsal root Identification of changes in gene expression in dor-
ganglia microenvironment of female BB Wistar sal root ganglia in diabetic neuropathy: correlation
diabetic rats with mild neuropathy. J Neurol Sci. with functional deficits. J Neuropathol Exp Neurol.
1994;127:36–42. 2006;65:722–32.
58. Zochodne DW, Ho LT. The influence of sulindac on 75. Price SA, Gardiner NJ, Duran-Jimenez B, et  al.
experimental streptozotocin-induced diabetic neu- Thioredoxin interacting protein is increased in sen-
ropathy. Can J Neurol Sci. 1994;21:194–202. sory neurons in experimental diabetes. Brain Res.
59. Zochodne DW, Nguyen C.  Increased peripheral 2006;1116(1):206–14.
nerve microvessels in early experimental diabetic 76. Chandrasekhar A, Komirishetty P, Areti A, et  al.
neuropathy: quantitative studies of nerve and dorsal Dual specificity phosphatases support axon plastic-
root ganglia. J Neurol Sci. 1999;166:40–6. ity and viability. Mol Neurobiol. 2021;58:391–407.
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 345

77. Cheng Y, Liu J, Luan Y, et al. Sarm1 gene deficiency 92. Mizisin AP, Powell HC, Myers RR.  Edema and
attenuates diabetic peripheral neuropathy in mice. increased endoneurial sodium in galactose neurop-
Diabetes. 2019;68:2120–30. athy. reversal with an aldose reductase inhibitor. J
78. Gerdts J, Summers DW, Milbrandt J, et al. Axon self-­ Neurol Sci. 1986;74:35–43.
destruction: new links among SARM1, MAPKs, and 93. Schmidt RE, Plurad SB, Sherman WR, et al. Effects
NAD+ metabolism. Neuron. 2016;89:449–60. of aldose reductase inhibitor sorbinil on neuroaxonal
79. Woo V, Cheng C, Duraikannu A, et al. Caspase-6 is dystrophy and levels of myo-inositol and sorbital in
a dispensable enabler of adult mammalian axonal symapthetic autonomic ganglia of streptozotocin-­
degeneration. Neuroscience. 2018;371:242–53. induced diabetic rats. Diabetes. 1989;38:569–79.
80. Mizisin AP, Powell HC.  Schwann cell changes 94. Sima AA, Bril V, Nathaniel V, et  al. Regeneration
induced as early as one week after galactose intoxi- and repair of myelinated fibers in sural-nerve
cation. Acta Neuropathol. 1997;93:611–8. biopsy specimens from patients with diabetic
81. Borghini I, Ania-Lahuerta A, Regazzi R, et  al. neuropathy treated with sorbinil. N Engl J Med.
Alpha, beta I, beta II, delta, and epsilon protein 1988;319:548–55.
kinase C isoforms and compound activity in the sci- 95. Zenon GJ, Abobo CV, Carter BL, et al. Potential use
atic nerve of normal and diabetic rats. J Neurochem. of aldose reductase inhibitors to prevent diabetic
1994;62:686–96. complications. Clin Pharm. 1990;9:446–57.
82. Brett FM, Kalichman MW, Calcutt NA, et al. Effects 96. Zherebitskaya E, Akude E, Smith DR, et  al.
of seven days of galactose feeding and aldose reduc- Development of selective axonopathy in adult
tase inhibition on mast cells and vessel morphometry sensory neurons isolated from diabetic rats: role
in rat sciatic nerve. J Neurol Sci. 1996;141:6–12. of glucose-induced oxidative stress. Diabetes.
83. Greene DA, Lattimer SA, Sima AA. Sorbitol, phos- 2009;58:1356–64.
phoinositides, and sodium-potassium-ATPase in the 97. Fernyhough P, Gallagher A, Averill SA, et  al.
pathogenesis of diabetic complications. N Engl J Duplicate use 14199 aberrant neurofilament phos-
Med. 1987;316:599–606. phorylation in sensory neurons of rats with diabetic
84. Gupta S, Sussman I, Mcarthur CS, et  al. neuropathy. Diabetes. 1999;48:881–9.
Endothelium-dependent inhibition of Na(+)-K+ 98. Huang TJ, Sayers NM, Verkhratsky A, et  al.
AtPase activity in rabbit aorta by hyperglycemia. Neurotrophin-3 prevents mitochondrial dysfunction
possible role of endothelium-derived nitric oxide. J in sensory neurons of streptozotocin-diabetic rats.
Clin Invest. 1992;90:727–32. Exp Neurol. 2005;194:279–83.
85. Mizisin AP, Li L, Calcutt NA.  Sorbitol accumu- 99. Low PA, Nickander KK, Tritschler HJ.  The roles
lation and transmembrane efflux in osmotically of oxidative stress and antioxidant treatment
stressed JS1 Schwannoma cells. Neurosci Lett. in experimental diabetic neuropathy. Diabetes.
1997;229:53–6. 1997;46:S38–42.
86. Roberts RE, Mclean WG.  Protein kinase C iso- 100. Ceriello A, Giugliano D.  Oxidative stress and dia-
zyme expression in sciatic nerves and spinal betic complications. In: Alberti KGGM, Zimmet P,
cords of experimentally diabetic rats. Brain Res. Defronzo RA, et al., editors. International textbook
1997;754:147–56. of diabetes mellitus. Chichester, New  York: Wiley;
87. Sima AAF, Brismar T, Yagihashi S.  Neuropathies 1997. p. 1453–61.
encountered in the spontaneously diabetic BB Wistar 101. Lyons TJ, Jenkins AJ.  Glycation, oxidation, and
rat. In: Dyck PJ, Thomas PK, Asbury AK, et al., edi- lipoxidation in the development of the complications
tors. Diabetic neuropathy. Toronto: W.B. Saunders; of diabetes: a carbonyl stress hypothesis. Diabetes
1987. Rev. 1997;5:365–91.
88. Cherian PV, Kamijo M, Angelides KJ, et al. Nodal 102. Zalba G, Beaumont J, San Jose G, et  al. Vascular
Na(+)-channel displacement is associated with oxidant stress: molecular mechanisms and patho-
nerve-conduction slowing in the chronically diabetic physiological implications. J Physiol Biochem.
BB/W rat: prevention by aldose reductase inhibition. 2000;56:57–64.
J Diabetes Complications. 1996;10:192–200. 103. Oates PJ.  Polyol pathway and diabetic peripheral
89. Ahlgren SC, Levine JD. Protein kinase C inhibitors neuropathy. Int Rev Neurobiol. 2002;50:325–92.
decrease hyperalgesia and C-fiber hyperexcitability 104. Chalk C, Benstead TJ, Moore F.  Aldose reduc-
in the streptozotocin-diabetic rat. J Neurophysiol. tase inhibitors for the treatment of diabetic
1994;72:684–92. polyneuropathy. Cochrane Database Syst Rev.
90. Ishii H, Jirousek MR, Koya D, et al. Amelioration of 2007;2007:Cd004572.
vascular dysfunctions in diabetic rats by an oral PKC 105. Apfel SC, Schwartz S, Adornato BT, et al. Efficacy
beta inhibitor. Science. 1996;272:728–31. and safety of recombinant human nerve growth factor
91. Judzewitsch RG, Jaspan JB, Polonsky KS, et  al. in patients with diabetic polyneuropathy: a random-
Aldose reductase inhibition improves nerve con- ized controlled trial. rhNGF Clinical Investigator
duction velocity in diabetic patients. N Engl J Med. Group. JAMA. 2000;284:2215–21.
1983;308:119–25. 106. Kessler JA, Smith AG, Cha BS, et al. Double-blind,
placebo-controlled study of HGF gene therapy
346 A. Areti and D. W. Zochodne

in diabetic neuropathy. Ann Clin Transl Neurol. vival by the PI3K-Akt signaling pathway. Curr Opin
2015;2:465–78. Neurobiol. 2001;11:297–305.
107. Frazier WA, Angeletti RH, Bradshaw RA.  Nerve 125. Namikawa K, Honma M, Abe K, et al. Akt/protein
growth factor and insulin. Science. 1972;176:482–8. kinase B prevents injury-induced motoneuron death
108. Ishii DN. Implication of insulin-like growth factors and accelerates axonal regeneration. J Neurosci.
in the pathogenesis of diabetic neuropathy. Brain 2000;20:2875–86.
Res Brain Res Rev. 1995;20:47–67. 126. Franke TF, Hornik CP, Segev L, et  al. PI3K/
109. Ishii DN. Insulin and related neurotrophic factors in Akt and apoptosis: size matters. Oncogene.
diabetic neuropathy. Diabet Med. 1993;10:14s–5s. 2003;22:8983–98.
110. Sugimoto K, Murakawa Y, Zhang W, et  al. Insulin 127. Hanada M, Feng J, Hemmings BA. Structure, regu-
receptor in rat peripheral nerve: its localization and lation and function of PKB/Akt—a major therapeu-
alternatively spliced isoforms. Diabetes Metab Res tic target. Biochim Biophys Acta. 2004;1697:3–16.
Rev. 2000;16:354–63. 128. Fernyhough P, Willars GB, Lindsay RM, et  al.
111. Sugimoto K, Murakawa Y, Sima AA.  Expression Insulin and insulin-like growth factor I enhance
and localization of insulin receptor in rat dorsal regeneration in cultured adult rat sensory neurones.
root ganglion and spinal cord. J Peripher Nerv Syst. Brain Res. 1993;607:117–24.
2002;7:44–53. 129. Toth C, Brussee V, Martinez JA, et  al. Rescue and
112. Fernyhough P, Mill JF, Roberts JL, et al. Stabilization regeneration of injured peripheral nerve axons by
of tubulin mRNAs by insulin and insulin-like growth intrathecal insulin. Neuroscience. 2006;139:429–49.
factor I during neurite formation. Brain Res Mol 130. Brussee V, Cunningham FA, Zochodne DW. Direct
Brain Res. 1989;6:109–20. insulin signaling of neurons reverses diabetic neu-
113. Chiu SL, Chen CM, Cline HT.  Insulin recep- ropathy. Diabetes. 2004;53:1824–30.
tor signaling regulates synapse number, dendritic 131. Singhal A, Cheng C, Sun H, et al. Near nerve local
plasticity, and circuit function in  vivo. Neuron. insulin prevents conduction slowing in experimental
2008;58:708–19. diabetes. Brain Res. 1997;763:209–14.
114. Havrankova J, Roth J, Brownstein M. Insulin recep- 132. Guo G, Kan M, Martinez JA, et  al. Local insulin
tors are widely distributed in the central nervous sys- and the rapid regrowth of diabetic epidermal axons.
tem of the rat. Nature. 1978;272:827–9. Neurobiol Dis. 2011;43:414–21.
115. Karagiannis SN, King RH, Thomas 133. Toth C, Brussee V, Zochodne DW.  Remote neuro-
PK. Colocalisation of insulin and IGF-1 receptors in trophic support of epidermal nerve fibres in experi-
cultured rat sensory and sympathetic ganglion cells. mental diabetes. Diabetologia. 2006;49:1081–8.
J Anat. 1997;191:431–40. 134. De La Hoz CL, Cheng C, Fernyhough P, et  al. A
116. Brussee V, Cunningham A, Zochodne DW.  Direct model of chronic diabetic polyneuropathy: benefits
insulin signalling of neurons reverses diabetic neu- from intranasal insulin are modified by sex and
ropathy. Diabetes. 2004;53:1824–30. RAGE deletion. Am J Physiol Endocrinol Metab.
117. Xu Q-G, Li X-Q, Kotecha SA, et al. Insulin as an 2017;312(5):E407–19.
in vivo growth factor. Exp Neurol. 2004;188:43–51. 135. Francis GJ, Martinez JA, Liu WQ, et  al. Motor
118. White MF. The insulin signalling system and the IRS end plate innervation loss in diabetes and the
proteins. Diabetologia. 1997;40:S2–17. role of insulin. J Neuropathol Exp Neurol.
119. Thirone AC, Huang C, Klip A. Tissue-specific roles 2011;70:323–39.
of IRS proteins in insulin signaling and glucose 136. Chen DK, Frizzi KE, Guernsey LS, et al. Repeated
transport. Trends Endocrinol Metab. 2006;17:72–8. monitoring of corneal nerves by confocal micros-
120. Heidenreich KA.  Insulin and IGF-I receptor sig- copy as an index of peripheral neuropathy in type-1
naling in cultured neurons. Ann N Y Acad Sci. diabetic rodents and the effects of topical insulin. J
1993;692:72–88. Peripher Nerv Syst. 2013;18:306–15.
121. Fadool DA, Tucker K, Phillips JJ, et  al. Brain 137. Cheng HL, Randolph A, Yee D, et al. Characterization
insulin receptor causes activity-dependent current of insulin-like growth factor-I and its receptor and
suppression in the olfactory bulb through mul-
­ binding proteins in transected nerves and cultured
tiple phosphorylation of Kv1.3. J Neurophysiol. Schwann cells. J Neurochem. 1996;66:525–36.
2000;83:2332–48. 138. Zochodne DW, Cheng C.  Neurotrophins and other
122. Folli F, Bonfanti L, Renard E, et al. Insulin receptor growth factors in the regenerative milieu of proximal
substrate-1 (IRS-1) distribution in the rat central ner- nerve stump tips. J Anat. 2000;196:279–83.
vous system. J Neurosci. 1994;14:6412–22. 139. Cheng HL, Russell JW, Feldman EL.  IGF-I pro-
123. Edstrom A, Ekstrom PA.  Role of phosphatidylino- motes peripheral nervous system myelination. Ann
sitol 3-kinase in neuronal survival and axonal out- N Y Acad Sci. 1999;883:124–30.
growth of adult mouse dorsal root ganglia explants. 140. Morino K, Petersen KF, Shulman GI.  Molecular
J Neurosci Res. 2003;74:726–35. mechanisms of insulin resistance in humans and
124. Brunet A, Datta SR, Greenberg ME. Transcription-­ their potential links with mitochondrial dysfunction.
dependent and -independent control of neuronal sur- Diabetes. 2006;55(Suppl 2):S9–S15.
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 347

141. Zeggini E, Parkinson J, Halford S, et al. Association tion across nerve transection gaps following a
studies of insulin receptor substrate 1 gene (IRS1) brief electrical stimulation paradigm. J Neurosurg.
variants in type 2 diabetes samples enriched for 2012;116:512.
family history and early age of onset. Diabetes. 158. Singh B, Krishnan A, Micu I, et al. Peripheral neuron
2004;53:3319–22. plasticity is enhanced by brief electrical stimulation
142. Esposito DL, Li Y, Vanni C, et  al. A novel T608R and overrides attenuated regrowth in experimental
missense mutation in insulin receptor substrate-1 diabetes. Neurobiol Dis. 2015;83:134–51.
identified in a subject with type 2 diabetes impairs 159. Geremia NM, Pettersson LM, Hasmatali JC, et  al.
metabolic insulin signaling. J Clin Endocrinol Endogenous BDNF regulates induction of intrinsic
Metab. 2003;88:1468–75. neuronal growth programs in injured sensory neu-
143. Imoto K, Kukidome D, Nishikawa T, et  al. Impact rons. Exp Neurol. 2010;223:128–42.
of mitochondrial reactive oxygen species and apop- 160. Kennedy JM, Zochodne DW. The regenerative defi-
tosis signal-regulating kinase 1 on insulin signaling. cit of peripheral nerves in experimental diabetes:
Diabetes. 2006;55:1197–204. its extent, timing and possible mechanisms. Brain.
144. Singh B, Xu Y, Guo GF, et al. Insulin signaling and 2000;123:2118–29.
insulin resistance in sensory neurons. J Peripher 161. Christie KJ, Webber CA, Martinez JA, et al. PTEN
Nerv Syst. 2009;13:254. inhibition to facilitate intrinsic regenerative out-
145. Singh B, Xu Y, McLaughlin T, et al. Resistance to tro- growth of adult peripheral axons. J Neurosci.
phic neurite outgrowth of sensory neurons exposed 2010;30:9306–15.
to insulin. J Neurochem. 2012;121(2):263–76. 162. Tucker BA, Rahimtula M, Mearow KM.  Laminin
146. Kim B, Mclean LL, Philip SS, et al. Hyperinsulinemia and growth factor receptor activation stimulates dif-
induces insulin resistance in dorsal root ganglion ferential growth responses in subpopulations of adult
neurons. Endocrinology. 2011;152:3638–47. DRG neurons. Eur J Neurosci. 2006;24:676–90.
147. Grote CW, Morris JK, Ryals JM, et al. Insulin recep- 163. Singh B, Singh V, Krishnan A, et al. Regeneration of
tor substrate 2 expression and involvement in neu- diabetic axons is enhanced by selective knockdown
ronal insulin resistance in diabetic neuropathy. Exp of the PTEN gene. Brain. 2014;137:1051–67.
Diabetes Res. 2011;2011:212571. 164. Pham VM, Tu NH, Katano T, et al. Impaired periph-
148. Kan M, Guo G, Singh B, et al. Glucagon-like peptide eral nerve regeneration in type-2 diabetic mouse
1, insulin, sensory neurons, and diabetic neuropathy. model. Eur J Neurosci. 2018;47:126–39.
J Neuropathol Exp Neurol. 2012;71:494–510. 165. Christie KJ, Krishnan A, Martinez JA, et  al.
149. Himeno T, Kamiya H, Naruse K, et  al. Beneficial Enhancing adult nerve regeneration through the
effects of exendin-4 on experimental polyneuropa- knockdown of retinoblastoma protein. Nat Commun.
thy in diabetic mice. Diabetes. 2011;60:2397–406. 2014;5:3670.
150. Jolivalt CG, Fineman M, Deacon CF, et  al. GLP-1 166. Futatsugi A, Utreras E, Rudrabhatla P, et al. Cyclin-­
signals via ERK in peripheral nerve and prevents dependent kinase 5 regulates E2F transcription
nerve dysfunction in diabetic mice. Diabetes Obes factor through phosphorylation of Rb protein in neu-
Metab. 2011;13:990–1000. rons. Cell Cycle. 2012;11:1603–10.
151. Bautista J, Chandrasekhar A, Komirishetty PK, et al. 167. Dimova DK, Dyson NJ. The E2F transcriptional net-
Regenerative plasticity of intact human skin axons. J work: old acquaintances with new faces. Oncogene.
Neurol Sci. 2020;417:117058. 2005;24:2810–26.
152. Senger JB, Verge VMK, Chan KM, et al. The nerve 168. Dick FA, Rubin SM. Molecular mechanisms under-
conditioning lesion: a strategy to enhance nerve lying Rb protein function. Nat Rev Mol Cell Biol.
regeneration. Ann Neurol. 2018;83:691–702. 2013;14:297–306.
153. Brushart TM, Hoffman PN, Royall RM, et  al. 169. Abe N, And Cavalli V. Nerve injury signaling. Curr
Electrical stimulation promotes motoneuron regen- Opin Neurobiol. 2008;18:276–83.
eration without increasing its speed or conditioning 170. Jones DM, Tucker BA, Rahimtula M, et  al. The
the neuron. J Neurosci. 2002;22:6631–8. synergistic effects of NGF and IGF-1 on neurite
154. Al-Majed AA, Neumann CM, Brushart TM, et  al. growth in adult sensory neurons: convergence on
Brief electrical stimulation promotes the speed and the PI 3-kinase signaling pathway. J Neurochem.
accuracy of motor axonal regeneration. J Neurosci. 2003;86:1116–28.
2000;20:2602–8. 171. Park KK, Liu K, Hu Y, et al. Promoting axon regen-
155. Brushart TM, Jari R, Verge V, et al. Electrical stimu- eration in the adult CNS by modulation of the PTEN/
lation restores the specificity of sensory axon regen- mTOR pathway. Science. 2008;322:963–6.
eration. Exp Neurol. 2005;194:221–9. 172. Culman J, Zhao Y, Gohlke P, et  al. PPAR-gamma:
156. Geremia NM, Gordon T, Brushart TM, et  al. therapeutic target for ischemic stroke. Trends
Electrical stimulation promotes sensory neuron Pharmacol Sci. 2007;28:244–9.
regeneration and growth-associated gene expression. 173. Bordet R, Ouk T, Petrault O, et  al. PPAR: a new
Exp Neurol. 2007;205:347–59. pharmacological target for neuroprotection in stroke
157. Singh B, Xu QG, Franz CK, et  al. Accelerated and neurodegenerative diseases. Biochem Soc Trans.
axon outgrowth, guidance, and target reinnerva- 2006;34:1341–6.
348 A. Areti and D. W. Zochodne

174. Iwata M, Haruta T, Usui I, et al. Pioglitazone ame- mitochondrial function and prevent neurodegenera-
liorates tumor necrosis factor-alpha-induced insulin tion. Mol Neurobiol. 2020;57:2521–38.
resistance by a mechanism independent of adipo- 190. Jolivalt CG, Frizzi KE, Han MM, et al. Topical deliv-
genic activity of peroxisome proliferator—activated ery of muscarinic receptor antagonists prevents and
receptor-gamma. Diabetes. 2001;50:1083–92. reverses peripheral neuropathy in female diabetic
175. Smith U.  Pioglitazone: mechanism of action. Int J mice. J Pharmacol Exp Ther. 2020;374:44–51.
Clin Pract Suppl. 2001;13–18. 191. Lee-Kubli CA, Calcutt NA.  Painful neuropathy:
176. Rieusset J, Auwerx J, Vidal H.  Regulation of mechanisms. Handb Clin Neurol. 2014;126:533–57.
gene expression by activation of the peroxisome 192. Greig M, Tesfaye S, Selvarajah D, et al. Insights into
proliferator-­activated receptor gamma with rosigli- the pathogenesis and treatment of painful diabetic
tazone (BRL 49653) in human adipocytes. Biochem neuropathy. Handb Clin Neurol. 2014;126:559–78.
Biophys Res Commun. 1999;265:265–71. 193. Wall PD, Devor M. Sensory afferent impulses origi-
177. Ribon V, Johnson JH, Camp HS, et  al. nate from dorsal root ganglia as well as from the
Thiazolidinediones and insulin resistance: peroxi- periphery in normal and nerve injured rats. Pain.
some proliferatoractivated receptor gamma activa- 1983;17:321–39.
tion stimulates expression of the CAP gene. Proc 194. Faber CG, Hoeijmakers JG, Ahn HS, et al. Gain of
Natl Acad Sci U S A. 1998;95:14751–6. function Na(V) 1.7 mutations in idiopathic small
178. Baumann CA, Chokshi N, Saltiel AR, et al. cloning fiber neuropathy. Ann Neurol. 2012;71:26–39.
and characterization of a functional peroxisome pro- 195. Lauria G, Ziegler D, Malik R, et  al. The role of
liferator activator receptor-gamma-responsive ele- sodium channels in painful diabetic and idiopathic
ment in the promoter of the CAP gene. J Biol Chem. neuropathy. Curr Diab Rep. 2014;14:538.
2000;275:9131–5. 196. Bierhaus A, Fleming T, Stoyanov S, et  al.
179. Duraikannu A, Martinez JA, Chandrasekhar A, Methylglyoxal modification of Nav1.8 facilitates
et al. expression and manipulation of the APC-beta-­ nociceptive neuron firing and causes hyperalgesia in
catenin pathway during peripheral neuron regenera- diabetic neuropathy. Nat Med. 2012;18:926–33.
tion. Sci Rep. 2018;8:13197. 197. Shah BS, Gonzalez MI, Bramwell S, et al. Beta3, a
180. Chitwood DH, Timmermans MC.  Small RNAs are novel auxiliary subunit for the voltage gated sodium
on the move. Nature. 2010;467:415–9. channel is upregulated in sensory neurones follow-
181. Chen YY, Mcdonald D, Cheng C, et  al. Axon and ing streptozocin induced diabetic neuropathy in rat.
Schwann cell partnership during nerve regrowth. J Neurosci Lett. 2001;309:1–4.
Neuropathol Exp Neurol. 2005;64:613–22. 198. Alsaloum M, Estacion M, Almomani R, et  al. A
182. Webber CA, Christie KJ, Cheng C, et al. Schwann gain-of-function sodium channel beta2-subunit
cells direct peripheral nerve regeneration through mutation in painful diabetic neuropathy. Mol Pain.
the Netrin-1 receptors, DCC and Unc5H2. Glia. 2019;15:1744806919849802.
2011;59:1503–17. 199. Todorovic SM, Lingle CJ. Pharmacological proper-
183. Verma P, Chierzi S, Codd AM, et  al. Axonal pro- ties of T-type Ca2+ current in adult rat sensory neu-
tein synthesis and degradation are necessary for rons: effects of anticonvulsant and anesthetic agents.
efficient growth cone regeneration. J Neurosci. J Neurophysiol. 1998;79:240–52.
2005;25:331–42. 200. Latham JR, Pathirathna S, Jagodic MM, et  al.
184. Willis DE, Twiss JL. The evolving roles of axonally Selective T-type calcium channel blockade alle-
synthesized proteins in regeneration. Curr Opin viates hyperalgesia in ob/ob mice. Diabetes.
Neurobiol. 2006;16:111–8. 2009;58:2656–65.
185. Zochodne DW, Allison JA, Ho W, et  al. Evidence 201. Todorovic SM, Jevtovic-Todorovic V.  Targeting of
for CGRP accumulation and activity in experimental Cav3.2 T-type calcium channels in peripheral sen-
neuromas. Am J Physiol. 1995;268:H584–90. sory neurons for the treatment of painful diabetic
186. Toth CC, Willis D, Twiss JL, et al. Locally synthe- neuropathy. Pflugers Arch. 2014;466:701–6.
sized calcitonin gene-related peptide has a critical 202. Orestes P, Osuru HP, Mcintire WE, et  al. Reversal
role in peripheral nerve regeneration. J Neuropathol of neuropathic pain in diabetes by targeting gly-
Exp Neurol. 2009;68:326–37. cosylation of Ca(V)3.2 T-type calcium channels.
187. Cheng C, Webber CA, Wang J, et  al. Activated Diabetes. 2013;62:3828–38.
RHOA and peripheral axon regeneration. Exp 203. Tsantoulas C, Lainez S, Wong S, et  al.
Neurol. 2008;212:358–69. Hyperpolarization-activated cyclic nucleotide-­
188. Calcutt NA, Smith DR, Frizzi K, et  al. Selective gated 2 (HCN2) ion channels drive pain in mouse
antagonism of muscarinic receptors is neuropro- models of diabetic neuropathy. Sci Transl Med.
tective in peripheral neuropathy. J Clin Invest. 2017;9:Eaam6072.
2017;127:608–22. 204. Zenker J, Poirot O, De Preux Charles AS, et  al.
189. Saleh A, Sabbir MG, Aghanoori MR, et  al. Altered distribution of juxtaparanodal Kv1.2 subunits
Muscarinic toxin 7 signals via Ca(2+)/calmodulin-­ mediates peripheral nerve hyperexcitability in type 2
dependent protein kinase kinase beta to augment diabetes mellitus. J Neurosci. 2012;32:7493–8.
Diabetic Sensory Neurons, Dorsal Root Ganglia, and Neuropathy 349

205. Andersson DA, Gentry C, Light E, et  al. 209. Zochodne DW, Verge VM, Cheng C, et  al. Nitric
Methylglyoxal evokes pain by stimulating TRPA1. oxide synthase activity and expression in experimen-
PLos One. 2013;8:E77986. tal diabetic neuropathy. J Neuropathol Exp Neurol.
206. Dull MM, Riegel K, Tappenbeck J, et  al. 2000;59:798–807.
Methylglyoxal causes pain and hyperalge- 210. Cheng C, Zochodne DW. Sensory neurons with acti-
sia in human through C-fiber activation. Pain. vated caspase-3 survive long-term experimental dia-
2019;160:2497–507. betes. Diabetes. 2003;52:2363–71.
207. Chowdhury SK, Smith DR, Fernyhough P. The role 211. Zochodne DW, Ho LT.  The influence of indo-
of aberrant mitochondrial bioenergetics in diabetic methacin and guanethidine on experimental strep-
neuropathy. Neurobiol Dis. 2013;51:56–65. tozotocin diabetic neuropathy. Can J Neurol Sci.
208. Brussee V, Guo GF, Dong YY, et al. Distal degenera- 1992;19:433–41.
tive sensory neuropathy in a long term type 2 diabe- 212. Van Der Sloot P, Mizisin A, Zochodne DW. Sulindac
tes rat model. Diabetes. 2008;57:1664–73. in established experimental diabetes: a follow-up
study. Can J Neurol Sci. 1995;22(3):198–201.
Micro- and Macrovascular Disease
in Diabetic Neuropathy

Lihong Chen and Aristidis Veves

1 Introduction Chronic diabetic complications are the result


of small vessel disease. Diabetic microangiopa-
Diabetes is often defined a “vascular disease” thy has been considered the main anatomic alter-
because of the early and extensive involvement ation leading to the development of retinopathy,
of the vascular tree observed in diabetic nephropathy, and neuropathy. Nevertheless, mac-
patients and, even, in those at risk of develop- roangiopathy, i.e., atherosclerosis of peripheral
ing diabetes. Both the micro- and macrocircu- arteries, is also a prominent feature of long-­
lation are affected, though the pathophysiology, lasting diabetes and is characterized as involving
histology, clinical history, and clinical sequelae predominantly distal arteries. The possible links
at the two vascular levels appear to be quite between diabetic micro- and macrovascular alter-
different. ations and nerve damage will be the focus of this
Historically, there have been two competing chapter.
hypotheses regarding the origins of diabetic neu-
ropathy. According to the first one, diabetic neu-
ropathy is secondary to the impairment of nerve 2 Microvascular Disease:
and Schwann cells, while the second one stated Overview and Anatomic
that diabetic neuropathy is mostly ascribed to Changes
microvascular disease. However, it is currently
realized that nerve and microvascular injury are Lesions specific for diabetes have been observed
both among the factors that contribute to nerve in the arterioles and capillaries of the foot and
dysfunction. other organs that are the typical targets of dia-

L. Chen
Division of Endocrinology, West China Hospital,
Sichuan University, Chengdu, China
A. Veves (*)
Department of Surgery, Beth Israel Deaconess
Medical Center, Boston, MA, USA
e-mail: aveves@bidmc.harvard.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 351
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_19
352 L. Chen and A. Veves

betic chronic complications. A nowadays-­ ity of the vessels further limiting their ability to
historical retrospective histological study dilate in response to different stimuli [12].
demonstrated the presence of PAS-positive mate- In the kidney, nonenzymatic glycosylation
rial in the arterioles of amputated limb specimens reduces the charge on the basement membrane,
from diabetic patients [1]. Though it was believed which may account for transudation of albumin,
for several years that the anatomic changes an expanded mesangium, and albuminuria [13].
described were occlusive in nature, in 1984 Similar increases in vascular permeability occur
LoGerfo and Coffmann recognized that, in dia- in the eye and probably contribute to macular
betic patients, there is no evidence of an occlu- exudate formation and retinopathy [14]. In sim-
sive microvascular disease [2]. Subsequent plest terms, microvascular structural alterations
prospective anatomic staining and arterial casting in diabetes result in an increased vascular perme-
studies have demonstrated the absence of an arte- ability and impaired autoregulation of blood flow
riolar occlusive lesion, thus dispelling the hope- and vascular tone.
less notion of diabetic “occlusive small vessel Previous studies have shown a correlation
disease” [3, 4]. between the development of diabetic chronic
While there is no occlusive lesion in the dia- complication and metabolic control with perhaps
betic microcirculation, other structural changes the strongest evidence coming from the Diabetes
do exist. Arteriolar hyalinization, corresponding Control and Complications Trial (DCCT), which
to the thickening of walls of arterioles, was enrolled type 1 diabetic patients, and the United
observed in patients with diabetes, regardless of Kingdom Prospective Diabetes Study (UKPDS),
hypertension [5]. These abnormalities are char- which enrolled type 2 diabetic patients [15, 16].
acterized by hypertrophic remodeling and are The results from both clinical trials clearly
associated with impaired endothelium-depen- showed a delay in the development and progres-
dent vasodilation in  vitro. Furthermore, endo- sion of retinopathy, nephropathy, and neuropathy
neurial capillary density was also found to be with intensive glycemic control, thus supporting
increased in patients with diabetes, suggesting the direct causal relationship between hypergly-
that ­capillary density may respond to diabetes- cemia and microcirculation impairment. In con-
induced nerve ischemia [6]. The thickening of trast, there was less evident for macrovascular
the capillary basement membrane, which is disease, which was assessed only in the UKPDS.
accompanied by pericyte degeneration and It should be emphasized hyperglycemia alone
endothelial cell hyperplasia, is the dominant is not sufficient to trigger generalized diabetic
structural change in diabetic neuropathy. The microvascular pathologies. The Joslin Diabetes
composition of basement membrane in diabetes Center 50-year Medalist Study has shown that
suggested that accumulation of glycosylated 30–35% patients with type 1 diabetes did not have
and cross-linked proteins contributed to the significant microvascular complications [17].
expanded membrane structure and impaired This suggests that there may be unidentified
function. This can affect endothelial cell and genetic or other endogenous protective factors
extracellular matrix interactions and represents that diminish the adverse microvascular effects of
a response to the metabolic changes related to hyperglycemia. It has been shown that multiple
diabetes and hyperglycemia [7–9]. Furthermore, mechanisms are involved in the adverse effects of
the thickness of basement membrane of skin hyperglycemia with vascular complications,
and nerve correlates with the extent of neuropa- including nonenzymatic glycation and the forma-
thy in diabetes [10]. However, this alteration tion of advanced glycation end products (AGEs);
does not lead to occlusion of the capillary enhanced reactive oxygen production and actions;
lumen, and arteriolar blood flow may be normal endoplasmic reticulum (ER) stress; and the acti-
or even increased despite these changes [11]. vation of the polyol pathway, the diacylglycerol
On the contrary, it might act as a barrier to the (DAG)–protein kinase C (PKC) pathway [5], Src
exchange of nutrients and/or increase the rigid- homology-2 domain-containing phosphatase-1
Micro- and Macrovascular Disease in Diabetic Neuropathy 353

Hyperglycemia

Genetic and
LocaI tissue
epigenetic
responses
modulators

Increase intracellular glucose metabolism

Cell signaling dysfunction Toxic metabolites Altered osmols and


(Activation of PKC, MAP kinase, (AGE, oxidants, redox potentiaI
SHP-1, and phosphatases) methylglyoxal (polyol pathway)

Oxidative stress, ER stress, Inflammatory cytokines


Loss of insulin actions Kallikrein-bradykinin activation

Vascular and specific tissue cells dysfunction and


abnormal turnover

Nephropathy Retinopathy Neuropathy

Fig. 1  Schema of hyperglycemia’s induced pathways to microvascular complications. MAP, mitogen-activated


protein

(SHP-1), and the reninangiotensin system (RAS) is the endothelium dependent and independent
and kallikrein-bradykinin (BK) systems (Fig. 1). vasodilation, in the skin microcirculation [18].
Although the structural alterations observed in Many glucose-related metabolic pathways can
diabetic capillaries do not affect the basal blood determine endothelium dysfunction: increased
flow, some functional abnormalities of the micro- aldose reductase activity leading to the imbal-
vascular circulation, which may eventually result ance in nicotinamide adenine dinucleotide
in a relative ischemia, have been extensively doc- phosphate (NADP)/nicotinamide adenine dinu-
umented. This aspect will be deeply discussed in cleotide phosphate, reduced form (NADPH);
the next section. auto-­oxidation of glucose leading to the forma-
tion of reactive oxygen species (ROS);
“advanced glycation end-products” (AGEs)
3 Pathophysiology produced by nonenzymatic glycation of pro-
of Microvascular Disease teins; abnormal n6-fatty acid metabolism and
and Endothelial Dysfunction inappropriate activation of protein kinase C. All
in Diabetes these different pathways lead to an increase of
oxidative stress which is responsible for a
3.1 Functional Changes reduced availability of nitric oxide and, in turn,
for a functional tissue hypoxia and the develop-
Diabetes mellitus, even in the absence of com- ment of diabetic chronic complications [19]
plications, impairs the vascular reactivity, that (Fig. 2).
354 L. Chen and A. Veves

DIABETES

Hyperglycemia

DAG Impaired
n-6 fatty acid Polyol Sugar autoxidation Advanced
metabolism pathway glycation

Triglycerides PKC
LDL

OXIDATIVE STRESS

ENDOTHELIAL DYSFUNCTION

Capillary blood flow endoneurial hypoxia

NERVE DYSFUNCTION

NCV, Regeneration, Structural

Fig. 2  New concepts in the pathogenesis of diabetic neuropathy

3.2 Microvascular Dysfunction els in skin from the foot of diabetic patients with
and Diabetic Neuropathy severe neuropathy and foot ulceration [21].
Differences in the microcirculation between
Microvascular reactivity is further reduced at the the foot and forearm levels have also been inves-
foot level in the presence of peripheral diabetic tigated, the main hypothesis being that increased
neuropathy. Endothelial nitric oxide synthase hydrostatic pressure in distal microcirculatory
(eNOS) is a key regulator of vascular nitric oxide beds, related to the orthostatic posture, affects the
production. Immunostaining of foot skin biopsies foot microcirculation more than at the forearm
in our unit, with antiserum to human eNOS, glu- level. The endothelium-dependent and -indepen-
cose transporter I (GLUT I), which is a functional dent vasodilation is, in fact, lower at the foot level
marker of the endothelium and von Willebrand when compared to the forearm in healthy sub-
factor, an anatomical marker, showed no differ- jects and both non-neuropathic and neuropathic
ence among diabetic patients with or without diabetic patients [22]. This forearm–foot gradient
peripheral neuropathy in the staining of GLUT I exists despite a similar baseline blood flow at the
and von Willebrand factor, while the staining for foot and forearm level. Therefore, it is reasonable
the eNOS was reduced in neuropathic patients to believe that erect posture may be a contribut-
(Fig.  3) [20]. Another study documented ing factor for the early development of the nerve
increased levels of iNOS and reduced eNOS lev- damage at the foot, compared to the forearm.
Micro- and Macrovascular Disease in Diabetic Neuropathy 355

Fig. 3  Expression of 80
eNOS in patients with
diabetic neuropathy
(black columns),
patients with both
diabetic neuropathy and 60
peripheral vascular
disease (hatched
columns) and healthy
subjects (white 40
columns). The
expression of eNOS was
reduced in both the
diabetic groups
compared to the healthy 20
subjects

0
absent reduced normal

Endoneurial microangiopathy was related to pulses. The final result is that despite no reduc-
the severity of neuropathy, suggesting that there tion in the total lower extremity blood flow, there
might be a causal relationship between impaired is functional ischemia that promoted injury and
microvasculature and diabetic neuropathy [6, impairs wound healing.
23]. In a large observational study, it was found
that endothelial function was a strong indepen-
dent predictor of DPN [24]. Experimental data 3.3 Autonomic Denervation
also showed that endothelium impairment could
cause neuropathy through involvement of the Autonomic nerve fibers take part in the modula-
Desert Hedgehog pathway [25]. tion of blood circulation. At rest, noradrenergic
sympathetic nerves are tonically active in normo-
3.2.1 Endothelium Dysfunction thermic environments and increase their activity
Previous studies have suggested that endothelium-­ during cold exposure, releasing both norepineph-
dependent vasodilation is impaired in diabetes, rine and co-transmitters to decrease skin blood
regardless of the presence of long-term compli- flow [30]. Autonomic neuropathy can compro-
cations [26–28]. It has also been shown that mise the diabetic microcirculation because of the
endothelium dysfunction precedes the develop- development of arterio-venous shunting due to
ment of macro- and microvascular diabetic com- sympathetic denervation. The opening of these
plications, indicating that endothelial impairment shunts leads to a maldistribution of blood between
could be a critical factor toward the development the nutritional capillaries and subpapillary ves-
of diabetic vascular complications [29]. However, sels, and consequent aggravation of microvascu-
these changes mainly involve the ability of lar ischemia. Studies employing sural nerve
microcirculation under stress, such as foot injury photography and fluorescein angiography as well
while they do not affect the baseline blood flow as other elegant techniques seem to support this
[20]. This feature, combined with the arteriove- concept [31, 32]. In addition to the above, these
nous shunt that is present at the foot level due to changes also result in impaired thermoregulation
autonomic neuropathy, can account for observa- [33].
tions in clinical practice, in which, diabetic neu- A loss of sympathetic tone is also responsible
ropathy with or without ulceration presents with for an increased capillary permeability in diabetic
a warm limb with distended veins and palpable patients with neuropathy [34, 35]. This might
356 L. Chen and A. Veves

Fig. 4  The nerve axon


reflex-related NERVE AXON REFLEX
vasodilation or
neurovascular response:
stimulation of the Acetylcholine
c-nociceptive nerve
fibers by acetylcholine
or other noxious stimuli
leads to antidromic
stimulation of the
adjacent c-fibers, which Nerve
secrete Calcitonin cell
Gene-Related Peptide
(CGRP) that causes
vasodilation and
increased local blood
flow
Vasodilation
CGRP

cause endoneurial edema, as demonstrated by % 300 286


employing magnetic resonance spectroscopy, 268
which can in turn represent another mechanism
leading to a reduction of endoneurial perfusion 200
and a worsening of the nerve damage [35]. The
increased lower extremity capillary pressure upon
assuming the erect posture, due to early loss of 100
postural vasoconstriction (mediated by the sympa- 41*
thetic fibers), might amplify this edematous effect. 29* 25*
0
DN DA DV DC C
3.4 Nerve Axon Reflex
Fig. 5  The neurovascular response (expressed as percent-
age of blood flow increase over the baseline blood flow) is
Diabetic somatic neuropathy can further affect significantly reduced at the foot level of diabetic patients
the skin microcirculation by the impairment of with peripheral somatic neuropathy (DN), autonomic neu-
the axon reflex-related vasodilatation (Lewis’ ropathy (DA), and peripheral artery disease (DV) com-
flare). Under normal conditions, the stimulus of pared to diabetic patients without complications (DC) and
healthy controls (C) *p < 0.001
the c-nociceptive nerve fibers not only travels in
the normal direction, centrally toward the spinal
cord, but also peripherally (“antidromic conduc- This neurovascular (N-V) response is signifi-
tion”) to local cutaneous blood vessels, causing a cantly reduced at the foot level in diabetic patients
vasodilatation by the release of vasoactive sub- with peripheral somatic neuropathy, autonomic
stances, such as calcitonin gene-related peptide neuropathy, and peripheral artery disease com-
(CGRP), Neuropeptide Y, substance P, and bra- pared to diabetic patients without complications
dykinin by the c fibers initiate neurogenic inflam- and healthy control subjects (Fig.  5) [22, 36].
mation (Fig. 4). This short circuit, or nerve axon Moreover, local anesthesia significantly reduces
reflex, is responsible for the Lewis’ triple flare the nerve axon reflex-related vasodilation at the
response to injury and plays an important role in foot of patients without peripheral neuropathy,
increasing local blood flow when it is mostly while it has no effect on the amount of the pre-­
needed, i.e., in condition of stress. anesthesia N-V vasodilation—which is already
Micro- and Macrovascular Disease in Diabetic Neuropathy 357

very low—at the foot of neuropathic patients The previous conclusions are supported by
[37]. This suggests that the main determinant of recent studies in experimental diabetes which
the presence of the neurovascular vasodilation is have demonstrated that epineurial arterioles of
c-fiber function and that its measurement could the sciatic nerve are innervated by sensory nerves
be used as a surrogate measure of the function of that contain CGRP and mediate a hyperemic
these fibers. response at this level [39]. Furthermore, it has
Neurovascular response is associated with been shown that in long-term diabetic rats the
measures of peripheral nerve function [37, 38]. amount of CGRP present in epineurial arterioles
Studies in our units have shown that an N-V is diminished, which could be due to a denerva-
response lower than 50% is highly sensitive tion process [40]. Exogenous CGRP-mediated
(90%) and adequately specific (74%), in identify- vasodilation of these arterioles is also impaired in
ing diabetic patients with peripheral neuropathy experimental diabetes, indicating a reduced
[38]. Besides, the finding that this response is sig- CGRP bioactivity [40]. All these findings further
nificantly reduced even in the early stages of support a role of small sensory nerve fibers’
peripheral neuropathy supports the hypothesis impairment in the development and progression
that small fiber damage is a precocious event in of diabetic neuropathy.
the clinical history of diabetic neuropathy—even The impairment of the nerve axon reflex-­
preceding large fibers’ impairment (Fig. 6). This related vasodilation is not affected by successful
leads to impaired vasodilation under conditions bypass surgery in patients with peripheral arterial
of stress, such as injury or inflammation. disease. In addition, the endothelium-dependent
Therefore, it is possible to speculate that small and -independent vasodilation, which is not
fiber neuropathy might further contribute to nerve related to the nerve axon reflex, remains impaired
hypoxic damage by the impairment of this hyper- after successful revascularization. Therefore,
emic response, determining a vicious cycle of despite correction in obstructive lesions and res-
injury. toration of normal blood flow in the large vessels,

180
% blood flow increase over baseline

*
160
*
140

120

100

80

60

40

20

0
C D DN mild DN moderate DN severe

Fig. 6  The nerve axon reflex-related vasodilation at the trols (C). Median (25–75 percentile). The nerve axon
foot level in a diabetic population stratified on the basis of reflex-related vasodilation is already significantly reduced
the degree of peripheral somatic neuropathy in patients in the early stages of neuropathy (subclinical neuropathy),
without neuropathy (D), with mild neuropathy (DN mild), supporting the belief that small fiber dysfunction might
with moderate neuropathy (DN moderate), and with precede large fiber impairment in the natural history of
severe neuropathy (DN severe) compared to healthy con- diabetic nerve damage
358 L. Chen and A. Veves

the changes in microcirculation continue to be than 50% patients with critical limb ischemia
present and cause tissue hypoxia under condi- having diabetes [47]. In addition, diabetes is also
tions of stress [41]. associated with more severe below-the-knee
In summary, both the functional and structural PAD.  The anterior tibial, posterior tibial, and
changes observed in diabetic microcirculation peroneal arteries are most commonly affected
contribute to the shift of blood flow away from and tend to have more diffuse lesions [48–50].
the nutritive capillaries to low-resistance arterio- Equally important is the observation that the
venous shunts, leading to functional ischemia of arteries of the foot, specifically the dorsalis
tissues including peripheral nerves and, conse- pedis, are often spared of occlusive disease. This
quently, to the development of diabetic periph- provides an excellent option for a distal revascu-
eral neuropathy and other diabetic chronic larization target [51].
complications. The clinical presentation and prognosis of
PAD with diabetes is also implicated by the coex-
istence of peripheral neuropathy. Microvascular
4 Lower Extremity Arterial disease (i.e., retinopathy, nephropathy, neuropa-
Disease and Diabetes thy) independently associated with limb amputa-
tion after controlling for traditional atherogenic
Smoking, diabetes mellitus, and dyslipidemia are risk factors. Concomitant presence of microvas-
the most associated risk factors of lower extrem- cular disease and PAD synergistically amplifies
ity artery disease [42]. Diabetes mellitus is esti- the risk of amputation. Thus, it is imperative to
mated to be associated with two- to fourfold take into consideration the importance of micro-
increase in the prevalence of peripheral arterial vascular disease in consideration of limb salvage
disease (PAD) [43]. The concomitant occurrence strategies [52]. In addition, diabetic patients are
of atherosclerotic peripheral vascular disease and less likely to present with typical claudication
peripheral neuropathy in patients with diabetes is symptoms than patients without diabetes. As a
the main factor in the development of diabetic consequence, the development of tissue loss (foot
foot pathology. Furthermore, peripheral arterial ulceration or gangrene) may represent the first
disease (PAD) of the lower extremities is consid- sign of lower limb ischemia and because of its
ered the major risk factor for lower-extremity limb-threatening potential, it is termed as critical
amputation, and it is also accompanied by a high limb ischemia. Therefore, diabetic patients with a
likelihood for cardiovascular and cerebrovascular foot ulcer should always be evaluated for isch-
diseases [44]. In a large, multinational emia, irrespective of their symptoms, particularly
­meta-­analysis, the presence of diabetes was asso- for the increased risk of limb-threatening infec-
ciated with nearly a twofold increased odds for tion and faulty healing related to PVD [53].
PAD [45]. The risk of amputation in patients with The observations that distal vessels (i.e., distal
diabetes is 0.2–2.0/1000, which is 10–15 times peroneal, dorsal pedal, and plantar arteries) are
greater in diabetes than in the nondiabetic popu- often spared from arterial occlusive disease had a
lation [46]. crucial impact on the manner in which peripheral
Although the underlying pathogenesis of ath- vascular disease is approached in the diabetic
erosclerotic disease in diabetics is similar to that population. In the past, based upon the false pre-
noted in nondiabetics, there are still some sig- sumption of small vessel disease, diabetics were
nificant differences. Diabetic patients present not treated as aggressively with revascularization
with atherosclerotic disease at a significantly as is now standard. A more aggressive attempt to
younger age than their nondiabetic counterparts, correct the vascular deficit in diabetic ischemic
tend to present with more advanced disease and limbs in addition to more aggressive measures to
have a worse prognosis. The prevalence of con- control local infection has radically altered the
comitant of PAD and diabetes is high in patients prognosis of peripheral vascular disease in the
with critical lower limb ischemia, with more diabetic extremity.
Micro- and Macrovascular Disease in Diabetic Neuropathy 359

5 Conclusions 10. Malik RA, Newrick PG, Sharma AK, Jennings A,


Ah-See AK, Mayhew TM, Jakubowski J, Boulton
AJ, Ward JD.  Microangiopathy in human diabetic
Changes in the micro- and macrocirculation, both neuropathy: relationship between capillary abnor-
anatomical and functional, contribute to the malities and the severity of neuropathy. Diabetologia.
development of diabetic neuropathy. On the other 1989;32:92–102.
11. Parving HH, Viberti GC, Keen H, Christiansen
hand, the development of diabetic neuropathy JS, Lassen NA.  Hemodynamic factors in the gen-
also affects the vasodilatory capacity of the esis of diabetic microangiopathy. Metabolism.
microcirculation and can interfere with the clini- 1983;32:943–9.
cal presentation of peripheral obstructive arterial 12. Flynn MD, Tooke JE.  Aetiology of diabetic foot
ulceration: a role for the microcirculation? Diabet
disease. Thus, the interaction between changes in Med. 1992;9:320–9.
the vasculature and peripheral nerves is bidirec- 13. Mogensen CE, Schmitz A, Christensen
tional and results in changes in both blood flow CK.  Comparative renal pathophysiology relevant to
and neuronal function. IDDM and NIDDM patients. Diabetes Metab Rev.
1988;4:453–83.
14. Cunha-Vaz JG.  Studies on the pathophysiology of
diabetic retinopathy. The blood-retinal barrier in dia-
References betes. Diabetes. 1983;32(Suppl 2):20–7.
15. Diabetes C, Complications Trial Research G, Nathan
1. Goldenberg S, Alex M, Joshi RA, Blumenthal DM, Genuth S, Lachin J, Cleary P, Crofford O, Davis
HT. Nonatheromatous peripheral vascular disease of M, Rand L, Siebert C.  The effect of intensive treat-
the lower extremity in diabetes mellitus. Diabetes. ment of diabetes on the development and progression
1959;8:261–73. of long-term complications in insulin-dependent dia-
2. LoGerfo FW, Coffman JD.  Current concepts. betes mellitus. N Engl J Med. 1993;329:977–86.
Vascular and microvascular disease of the foot in 16. Group UPDSU.  Intensive blood-glucose control
diabetes. Implications for foot care. N Engl J Med. with sulphonylureas or insulin compared with con-
1984;311:1615–9. ventional treatment and risk of complications in
3. Conrad MC. Large and small artery occlusion in dia- patients with type 2 diabetes (UKPDS 33). Lancet.
betics and nondiabetics with severe vascular disease. 1998;352:837–53.
Circulation. 1967;36:83–91. 17. Keenan HA, Costacou T, Sun JK, Doria A, Cavellerano
4. Strandness DE Jr, Priest RE, Gibbons GE. Combined J, Coney J, Orchard TJ, Aiello LP, King GL. Clinical
clinical and pathologic study of diabetic and non- factors associated with resistance to microvascular
diabetic peripheral arterial disease. Diabetes. complications in diabetic patients of extreme disease
1964;13:366–72. duration: the 50-year medalist study. Diabetes Care.
5. Rizzoni D, Porteri E, Guelfi D, Muiesan ML, 2007;30:1995–7.
Valentini U, Cimino A, Girelli A, Rodella L, Bianchi 18. Morris SJ, Shore AC, Tooke JE.  Responses of the
R, Sleiman I, Rosei EA. Structural alterations in sub- skin microcirculation to acetylcholine and sodium
cutaneous small arteries of normotensive and hyper- nitroprusside in patients with NIDDM. Diabetologia.
tensive patients with non-insulin-dependent diabetes 1995;38:1337–44.
mellitus. Circulation. 2001;103:1238–44. 19. Cameron NE, Eaton SE, Cotter MA, Tesfaye
6. Thrainsdottir S, Malik RA, Dahlin LB, Wiksell S. Vascular factors and metabolic interactions in the
P, Eriksson KF, Rosen I, Petersson J, Greene DA, pathogenesis of diabetic neuropathy. Diabetologia.
Sundkvist G.  Endoneurial capillary abnormali- 2001;44:1973–88.
ties presage deterioration of glucose tolerance and 20. Veves A, Akbari CM, Primavera J, Donaghue VM,
accompany peripheral neuropathy in man. Diabetes. Zacharoulis D, Chrzan JS, DeGirolami U, LoGerfo
2003;52:2615–22. FW, Freeman R.  Endothelial dysfunction and the
7. LeBleu VS, Macdonald B, Kalluri R.  Structure and expression of endothelial nitric oxide synthetase in
function of basement membranes. Exp Biol Med. diabetic neuropathy, vascular disease, and foot ulcer-
2007;232:1121–9. ation. Diabetes. 1998;47:457–63.
8. Haitoglou CS, Tsilibary EC, Brownlee M, Charonis 21. Jude EB, Boulton AJ, Ferguson MW, Appleton I. The
AS. Altered cellular interactions between endothelial role of nitric oxide synthase isoforms and arginase
cells and nonenzymatically glucosylated laminin/type in the pathogenesis of diabetic foot ulcers: possible
IV collagen. J Biol Chem. 1992;267:12404–7. modulatory effects by transforming growth factor
9. Williamson JR, Tilton RG, Chang K, Kilo beta 1. Diabetologia. 1999;42:748–57.
C.  Basement membrane abnormalities in diabetes 22. Arora S, Smakowski P, Frykberg RG, Simeone LR,
mellitus: relationship to clinical microangiopathy. Freeman R, LoGerfo FW, Veves A.  Differences in
Diabetes Metab Rev. 1988;4:339–70. foot and forearm skin microcirculation in diabetic
360 L. Chen and A. Veves

patients with and without neuropathy. Diabetes Care. 37. Caselli A, Rich J, Hanane T, Uccioli L, Veves
1998;21:1339–44. A.  Role of C-nociceptive fibers in the nerve axon
23. Malik RA, Tesfaye S, Thompson SD, Veves A, reflex-related vasodilation in diabetes. Neurology.
Sharma AK, Boulton AJ, Ward JD. Endoneurial local- 2003;60:297–300.
isation of microvascular damage in human diabetic 38. Caselli A, Spallone V, Marfia GA, Battista C, Pachatz
neuropathy. Diabetologia. 1993;36:454–9. C, Veves A, Uccioli L.  Validation of the nerve axon
24. Roustit M, Loader J, Deusenbery C, Baltzis D, Veves reflex for the assessment of small nerve fibre dysfunc-
A. Endothelial dysfunction as a link between cardio- tion. J Neurol Neurosurg Psychiatry. 2006;77:927–32.
vascular risk factors and peripheral neuropathy in 39. Calcutt NA, Chen P, Hua XY. Effects of diabetes on
diabetes. J Clin Endocrinol Metab. 2016;101:3401–8. tissue content and evoked release of calcitonin gene-­
25. Chapouly C, Yao Q, Vandierdonck S, Larrieu-Lahargue related peptide-like immunoreactivity from rat sen-
F, Mariani JN, Gadeau AP, Renault MA.  Impaired sory nerves. Neurosci Lett. 1998;254:129–32.
Hedgehog signalling-induced endothelial dysfunction 40. Yorek MA, Coppey LJ, Gellett JS, Davidson
is sufficient to induce neuropathy: implication in dia- EP. Sensory nerve innervation of epineurial arterioles
betes. Cardiovasc Res. 2016;109:217–27. of the sciatic nerve containing calcitonin gene-related
26. Williams SB, Cusco JA, Roddy MA, Johnstone MT, peptide: effect of streptozotocin-induced diabetes.
Creager MA. Impaired nitric oxide-mediated vasodi- Exp Diabetes Res. 2004;5:187–93.
lation in patients with non-insulin-dependent diabetes 41. Arora S, Pomposelli F, LoGerfo FW, Veves
mellitus. J Am Coll Cardiol. 1996;27:567–74. A. Cutaneous microcirculation in the neuropathic dia-
27. Palmer RM, Ashton DS, Moncada S. Vascular endo- betic foot improves significantly but not completely
thelial cells synthesize nitric oxide from L-arginine. after successful lower extremity revascularization. J
Nature. 1988;333:664–6. Vasc Surg. 2002;35:501–5.
28. Johnstone MT, Creager SJ, Scales KM, Cusco JA, Lee 42. Criqui MH, Aboyans V.  Epidemiology of peripheral
BK, Creager MA.  Impaired endothelium-dependent artery disease. Circ Res. 2015;116:1509–26.
vasodilation in patients with insulin-dependent diabe- 43. Beckman JA, Creager MA, Libby P.  Diabetes and
tes mellitus. Circulation. 1993;88:2510–6. atherosclerosis: epidemiology, pathophysiology, and
29. Elhadd TA, Kennedy G, Hill A, McLaren M, Newton management. JAMA. 2002;287:2570–81.
RW, Greene SA, Belch JJ.  Abnormal markers of 44. Reiber GE, Pecoraro RE, Koepsell TD. Risk factors for
endothelial cell activation and oxidative stress in chil- amputation in patients with diabetes mellitus. A case-­
dren, adolescents and young adults with type 1 diabe- control study. Ann Intern Med. 1992;117:97–105.
tes with no clinical vascular disease. Diabetes Metab 45. Fowkes FG, Rudan D, Rudan I, Aboyans V,
Res Rev. 1999;15:405–11. Denenberg JO, McDermott MM, Norman PE,
30. Charkoudian N. Mechanisms and modifiers of reflex Sampson UK, Williams LJ, Mensah GA, Criqui
induced cutaneous vasodilation and vasoconstriction MH. Comparison of global estimates of prevalence
in humans. J Appl Physiol. 2010;109:1221–8. and risk factors for peripheral artery disease in 2000
31. Tesfaye S, Harris N, Jakubowski JJ, Mody C, Wilson and 2010: a systematic review and analysis. Lancet.
RM, Rennie IG, Ward JD.  Impaired blood flow and 2013;382:1329–40.
arterio-venous shunting in human diabetic ­neuropathy: 46. Vadiveloo T, Jeffcoate W, Donnan PT, Colhoun
a novel technique of nerve photography and fluores- HC, McGurnaghan S, Wild S, McCrimmon R,
cein angiography. Diabetologia. 1993;36:1266–74. Leese GP.  Scottish Diabetes Research Network
32. Flynn MD, Tooke JE.  Diabetic neuropathy and the Epidemiology G. Amputation-free survival in 17,353
microcirculation. Diabet Med. 1995;12:298–301. people at high risk for foot ulceration in diabe-
33. Rutkove SB, Veves A, Mitsa T, Nie R, Fogerson PM, tes: a national observational study. Diabetologia.
Garmirian LP, Nardin RA.  Impaired distal thermo- 2018;61:2590–7.
regulation in diabetes and diabetic polyneuropathy. 47. Dick F, Diehm N, Galimanis A, Husmann M,
Diabetes Care. 2009;32:671–6. Schmidli J, Baumgartner I. Surgical or endovascular
34. Lefrandt JD, Bosma E, Oomen PH, Hoeven JH, Roon revascularization in patients with critical limb isch-
AM, Smit AJ, Hoogenberg K. Sympathetic mediated emia: influence of diabetes mellitus on clinical out-
vasomotion and skin capillary permeability in diabetic come. J Vasc Surg. 2007;45:751–61.
patients with peripheral neuropathy. Diabetologia. 48. Jude EB, Oyibo SO, Chalmers N, Boulton
2003;46:40–7. AJ. Peripheral arterial disease in diabetic and nondia-
35. Eaton RP, Qualls C, Bicknell J, Sibbitt WL Jr, King betic patients: a comparison of severity and outcome.
MK, Griffey RH.  Structure-function relationships Diabetes Care. 2001;24:1433–7.
within peripheral nerves in diabetic neuropathy: the 49. American Diabetes A.  Peripheral arterial dis-
hydration hypothesis. Diabetologia. 1996;39:439–46. ease in people with diabetes. Diabetes Care.
36. Hamdy O, Abou-Elenin K, LoGerfo FW, Horton ES, 2003;26:3333–41.
Veves A.  Contribution of nerve-axon reflex-related 50. Armstrong EJ, Waltenberger J, Rogers
vasodilation to the total skin vasodilation in diabetic JH.  Percutaneous coronary intervention in patients
patients with and without neuropathy. Diabetes Care. with diabetes: current concepts and future directions.
2001;24:344–9. J Diabetes Sci Technol. 2014;8:581–9.
Micro- and Macrovascular Disease in Diabetic Neuropathy 361

51. Van Damme H. Crural or pedal artery revascularisa- 53. Weitz JI, Byrne J, Clagett GP, Farkouh ME, Porter JM,
tion for limb salvage: is it justified? Acta Chir Belg. Sackett DL, Strandness DE Jr, Taylor LM. Diagnosis
2004;104:148–57. and treatment of chronic arterial insufficiency of
52. Behroozian A, Beckman JA.  Microvascular dis- the lower extremities: a critical review. Circulation.
ease increases amputation in patients with periph- 1996;94:3026–49.
eral artery disease. Arterioscler Thromb Vasc Biol.
2020;40:534–40.
The Spinal Cord in Diabetic
Neuropathy

Andrew G. Marshall, Anne Worthington,
and Corinne G. Jolivalt

1 Introduction flow of afferent information from the periphery


to the higher central nervous system, it is increas-
The spinal cord role in diabetic neuropathy has ingly evident that the spinal cord is a complex
been described since early 1900 and reviewed in sensorimotor processing interface. The spinal
detail in the previous edition [1]. In contrast to cord is the first site of integration of sensory input
the vulnerable peripheral nerves, the spinal cord from the periphery and the last site of descending
is exposed to less glycemic stress as a conse- control of sensory and motor systems; therefore,
quence of lower glucose levels in the cerebral disruption of its function may impede appropri-
spinal fluid than in the plasma [2]. However, the ate CNS control systems and contribute to appar-
spinal cord is not protected from diabetes-­ ent peripheral neuropathy [5].
induced injury. Indeed, structural and functional The number of studies on the function and
damage are discernable in the spinal cord of dia- contribution of the spinal cord to diabetes neu-
betic patients with histological evidence of neu- ropathy has more than trippled in the last 15 years
ropathy, radiculopathy, and myelopathy [1] and (from 15  articles/year in 2006 to 45–50/year in
clear atrophy detectable by magnetic resonance the last 5 years for spinal cord and diabetic neu-
imaging (MRI) scans [3, 4]. Structural damages ropathy in PubMed search). The main progress
are also detected in the spinal cord of animal areas in the last 15 years will be developed in this
models of diabetes, although not to the extent of chapter.
structural degeneration seen in human [1]. Rather
than merely acting as a passive conduit for the • Inflammation
• Molecular mechanisms
A. G. Marshall • Spinal disinhibition
Musculoskeletal Biology, Institute of Life Course and • Spinal cord stimulation
Medical Sciences, University of Liverpool,
Liverpool, UK
A. Worthington 2 Inflammation
Division of Diabetes, Endocrinology and
Gastroenterology, School of Medical Sciences,
Faculty of Biology, Medicine and Health, University 2.1 Microglia Activation in Type 1
of Manchester, Manchester, UK Diabetes
C. G. Jolivalt (*)
University of California San Diego, Several mechanisms taking place at the spinal cord
La Jolla, CA, USA level may underline the development and mainte-
e-mail: cjolivalt@ucsd.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 363
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_20
364 A. G. Marshall et al.

nance of diabetic peripheral neuropathy. Although cord, a phenomenon which comprises various
diabetic neuropathy is not an inflammatory neu- pathophysiological mechanisms responsible for
ropathy per se, among these mechanisms, activa- neuropathic pain-like signs in animal models.
tion of microglia and subsequent release of NMDAR function is modulated by post-transla-
proinflammatory cytokines and reactive oxygen tional modifications including phosphorylation,
intermediates have been suggested as playing an and this is proposed to underlie its involvement in
important role in spinal sensitization. Systemic the production of pain hypersensitivity. In particu-
changes such as hyperglycemia may be involved lar, increased phosphorylation of NMDAR1 was
in microglial activation. Under culture conditions, found in neurons and microglial cells in the spinal
hyperglycemia can cause morphological changes cord of STZ rats developing hyperalgesia.
indicative of activation of microglia as well as Blockers of MEK (an upstream kinase of ERK)
increased release of inflammatory cytokines [6, 7]. and of NMDAR suppressed hyperalgesia and
However, hyperglycemia was shown to be neces- decreased NMDAR phosphorylation, demonstrat-
sary but not sufficient to induce hyperalgesia, ing involvement of neuronal and microglial
which was associated with increase of extracellu- NMDA and ERK in central sensitization and
lar signal-regulated kinase (ERK), cJun N-terminal mechanical hyperalgesia in diabetes [16].
kinase (JNK) and p38 phosphorylation in the spi- Microglia involvement in allodynia in type 1
nal cord and dorsal root ganglia (DRG), more par- diabetic rats was further supported by adminis-
ticularly in neurons and microglia of streptozotocin tration of gabapentin or minocycline, which
(STZ) diabetic rats [8]. Microglial density attenuated microglial activation (reduction of
increased along with phosphorylated p38 MAPK expression of the early gene cFos) that correlated
(a marker of microglial activation) in diabetic with reduced allodynia in STZ diabetic rats [13,
rodent spinal cord, correlating with mechanical 14, 17–19]. Inhibition of microglial activation by
hyperalgesia [9, 10], and a cannabinoid receptor minocycline restored normal spinal expression of
B2 (CB2) agonist attenuated the neuropathic state, OX-42, BDNF, and DREAM (a calcium regu-
with reduced elevation of microglial density and lated transcriptional repressor) proteins [20].
phosphorylated p38  in diabetic mice [10]. Increased microglial activation and protein
Similarly, exercise, gabapentin, and lidocaine expression of BDNF and DREAM were not pres-
attenuated microglia activation, as measured by ent in diabetic rats without painful neuropathy
p38 phosphorylation in diabetic rodents [11–13]. (hypoalgesia after formalin injection) [21].
However, in our group, we have shown that differ- Peripheral infiltrated macrophage may also
ences exist between microglial activation observed contribute to the spinal cord inflammation as their
in diabetic rats and that observed in nerve-ligated number increased throughout the development of
rats (Dr. Lee-Kubli, unpublished). Unlike nerve diabetic neuropathy in STZ mice. Administration
injury that caused a large increase in the total num- of clodronate liposomes to deplete monocyte
ber of microglia, diabetes was not associated with reduced infiltrated macrophages along with alle-
a change in microglia number. However, the per- viation of tactile allodynia [22].
centage of microglia with retracted processes was Similarly, alteration of astrocytes activation
similar in both diabetic and nerve-ligated rats and may be involved in diabetic neuropathic pain.
was consistent with descriptions of morphological However, the role of astrogliosis in the hyperal-
activation depicted in the literature [8, 14, 15]. gesia detected in type 1 diabetic rodents is not
Additionally, the mitogen-activated protein clear, with report showing decreased GFAP-­
kinase (MAPK) phosphorylation was not detected positive astrocytes [23, 24] while other reported
in diabetic non-hyperalgesic rats, and N-methyl- increased activation of astrocytes in addition to
D-aspartate receptor (NMDAR) activation was microglial activation in the spinal cord of STZ
required for the MAPK phosphorylation in neu- diabetic rats [25]. Reduced activation of microg-
rons and microglia and consequent hyperalgesia lia and astrogliosis were also observed in the spi-
in the diabetic hyperalgesic rats [8]. The NMDAR nal cord of the HFD/STZ diabetes model only at
contributes to central sensitization in the spinal the late stage (after 30 days of diabetes) [26].
The Spinal Cord in Diabetic Neuropathy 365

2.2 Proinflammatory Peptides to reduce microglial activation and cytokines


and Cytokines release are reported in Table 1.
Kinins are defined as pro-inflammatory and
Various studies have shown increased proinflam- vasoactive peptides, which act through the activa-
matory cytokines levels in diabetic rodent spinal tion of two G-protein-coupled receptors denoted
cord. Here we report only recent studies for as B1 and B2 [42, 43]. The pro-nociceptive kinin
kinins and TNFα, while studies showing efficacy B1 receptor (B1R) is upregulated on sensory

Table 1  Studies showing efficacy on microglial activation and proinflammatory cytokines


Efficacy on neuropathic
Compound, extract, drug (main Animal pain indices [not assessed
function) Reduction of levels model (NA)] References
Curcumin (antioxidant) TNFα, TNFα receptor 1 STZ Mechanical allodynia [27]
diabetic and thermal hyperalgesia
rats
Puerarin (Chinese remedy) Microglia and astrocytes STZ Mechanical and thermal [28]
expression, NFκB, Il1β, diabetic nociceptive responses
IL6, TNFα rats
Gabapentin (anticonvulsant) Neutrophils, l-selectin STZ Allodynia and thermal [29]
(only one among 29 diabetic hyperalgesia
cytokines) rats
Hydralazine (scavenger of Microglia activation, Il1β, STZ Mechanical allodynia [30]
Acrolein, a lipid peroxidation TNFα diabetic and thermal hyperalgesia
product) rats
Danggui Sini decoction Microglia and astrocytes STZ Allodynia and thermal [31]
activation, Il1β, IL6, diabetic hyperalgesia
TNFα, NFκΒ rats
Ammoxetine (SNRI) Microglia activation, Il1β, STZ Mechanical allodynia [32]
IL6, TNFα diabetic
rats
Dexmedetomidine (sedative, Il1β, TNFα STZ Mechanical and thermal [33]
analgesic) diabetic nociceptive responses
rats
GS-KG9-Ginseng Microglia activation STZ Mechanical and thermal [34]
diabetic nociceptive responses
rats
Gabapentin, Levetiracetam Microglia and astrocytes STZ Allodynia and thermal [35]
(anticonvulsant) expression diabetic hyperalgesia
mice
Bone marrow-derived Microglia and astrocytes STZ Mechanical allodynia [36]
mesenchymal stem cells activation, Il1β, TNFα diabetic and thermal hypoalgesia
mice
Tramadol (opiate) and Il1β STZ Thermal hyperalgesia [37]
gabapentin diabetic
mice
Duloxetine (SSRI) Microglia and astrocytes Alloxan Allodynia and thermal [38]
expression diabetic hyperalgesia
mice
Valproate sodium (antiepileptic) Il1β, TNFα Alloxan Allodynia and thermal [39]
diabetic hyperalgesia
mice
Salidroside (anti-inflammatory Il1β, TNFα ZDF Mechanical allodynia [40]
phenylpropanoid glucoside) diabetic and thermal hyperalgesia
rats
MK801 (NMDAR antagonist) IL6, TNFα Db/db mice Allodynia and thermal [41]
hyperalgesia
366 A. G. Marshall et al.

C-fibers, astrocytes, and microglia in the spinal sion of IL1β and increased phosphorylation of
cord of STZ diabetic rat. This upregulation was NMDAR in spinal dorsal horn neurons of type 2
associated with increased Iba1 staining and diabetic mice [49]. In type 2 diabetic mice, astro-
reversed by microglia inhibitors (fluorocitrate cytic inhibition attenuated allodynia while
and minocycline), while improving tactile allo- microglia inhibition had no effect [49].
dynia [44]. Phosphorylated ERK was increased in the spi-
Tumor necrosis factor alpha (TNFα) is a pro- nal cord and DRG of type 2 diabetic mice (db/db)
inflammatory cytokine that has been implicated [50] and inhibition of MEK (ERK Kinase) atten-
as a key pain mediator in the development and uated mechanical allodynia, thermal hyperalge-
maintenance of neuropathic pain conditions. sia, and formalin response in these mice [51].
TNFα was significantly increased in STZ dia- The enhanced ERK phosphorylation was detected
betic rats that had painful diabetic neuropathy in projection sensory neurons and was associated
and to a lesser extent in diabetic rats with non-­ with astrocyte activation, increased NMDAR1
painful neuropathy as determined by their phosphorylation in projection neurons and upreg-
response to von Frey filaments [45]. The produc- ulation of nitric oxide synthase (neuronal and
tion of TNFα in the spinal cord has been shown to inducible) in interneurons and astrocytes peaking
be increased in diabetic mice, and attenuation of up at 10 weeks of diabetes [50]. Inhibition of
this overproduction by a selective cannabinoid NMDAR, MAPK, or NOS decreased ERK phos-
CB1 receptor antagonist prevented mechanical phorylation, NOS upregulation, and reduced
allodynia [46]. Systemic or intrathecal adminis- astrocytosis and GFAP upregulation in db/db
tration of etanercept, a TNFα inhibitor, alleviated mice spinal cord while inhibiting mechanical
tactile allodynia in diabetic mice [47]. The allodynia [50]. Increased ERK activation was
increased TNFα levels in diabetic rat spinal cord demonstrated in ZDF rats following pressure of
was also reduced by treatment with a Poly(ADP-­ the hindpaw. This was reduced after 7 weeks of
ribose)Polymerase-1 (PARP) inhibitor [48]. treatment with pioglitazone (a peroxisome
TNFα has been shown to be elevated in diabetic proliferator-­activated receptor γ (PPARγ) ligand)
spinal cord, and its modulation improved painful [52]. However, the treatment reversed hypogly-
diabetic neuropathy. cemia, therefore the effect on spinal sensitization
Numerous studies have shown activation of cannot be attribute to pioglitazone alone but par-
microglia and release of proinflammatory cyto- tially to a reversal of diabetes.
kines (Il1β, TNFα) in STZ rodents that were
reduced by administration of various compounds,
extracts, and drugs (Table 1). 3 Molecular Mechanisms
Modulation of spinal cord inflammation in
rodent models of diabetic neuropathy supports 3.1 Mitochondrial Function
the contribution of inflammation to painful dia-
betic neuropathy and shows it could be a benefi- Mitochondrial respiratory chain activity is
cial therapeutical avenue to explore for treatment reduced in heart, kidney, skeletal muscle, and
of diabetic neuropathy. DRG of patients with type 2 diabetes and/or dia-
betic rodents (reviewed in [53]) and contributes
to impaired bioenergetics due to alteration of the
2.3 Inflammation in Type 2 AMP-activated protein kinase (AMPK)/sirtuin
Diabetes (SIRT)/peroxisome proliferator-activated recep-
tor γ coactivator α (PGC1α) pathway. In contrast
In contrast to type 1 diabetic rodent models, spi- to the peripheral nerves and the brain [53–55],
nal astrocytes, but not microglia, were shown to little is known about mitochondria dynamics, dis-
be activated in type 2 diabetic mice. The astro- tribution, and function in the diabetic spinal cord.
cyte activation correlated with increased expres- Mitochondrial fission was increased, resulting in
The Spinal Cord in Diabetic Neuropathy 367

the reduction of mitochondria in neuronal axons, development and structural plasticity of dendrites
and was accompanied by an abnormal distribu- and dendritic spines [64, 65]. Several studies
tion of mitochondria away from the axons toward have suggested that RhoA regulates the stability
the cell bodies in the spinal cord neurons of type of dendritic branches and spines in neurons [64,
1 diabetic rats and in ventral spinal cord neurons 66]. High glucose levels can induce the activation
in culture in high glucose condition [56]. The of RhoA through multiple mechanisms mediated
mitochondria also appeared fragmented. Another by PKC, cyclic guanosine monophosphatase
recent study showed reduction in mitochondrial (cGMP)-dependent protein kinase G (PKG), and
mass in the STZ diabetic rat spinal cord [57, 58]. reactive oxygen species [67]. It was also shown
More studies on spinal cord mitochondria are that RhoA and its downstream kinase, Rho kinase
required to fully characterize the role of the spi- (ROCK), play important roles in the development
nal cord in diabetic neuropathy. and/or maintenance of chronic pain [68, 69], and
the direct activation of ROCK is involved in dia-
betic painful neuropathy. The expression of
3.2 Synaptic Plasticity cleaved product of ROCK was increased in rats
fed a high-fructose diet [70]. Furthermore, ROCK
In contrast to the spinal mitochondria, synaptic inhibitors have shown efficacy in STZ diabetic
plasticity has been extensively studied in the last rat with attenuation of mechanical allodynia and
15 years. thermal responses, and reduction of WDR neuron
Imaging of the spinal cord of type 1 diabetic hyperexcitability [61, 71]. Other intracellular
rats by BOLD functional MRI demonstrated a pathways and proteins play a role in the altered
decreased blood-oxygen-level-dependent activ- synaptic plasticity in diabetes. APPL1, a neuro-
ity, indicative of changes in synapses between nal adaptor protein, affects synaptic plasticity,
primary afferents and second order neurons [59]. but its defined role in the pathogenesis of painful
Synaptic and dendritic alterations were detected diabetic neuropathy is under investigation.
after established diabetes in type 1 diabetic rats APPL1 expression was decreased in neurons and
with reduced axonal stabilization and dendritic microglia of diabetic rats that presented mechani-
structures associated with decreased expression cal and thermal hyperalgesia. Reduced APPL1
of ILK-PINCH proteins levels in the spinal cord was associated with activation of mTor, reduction
[60]. In STZ diabetic rats, alterations in dendritic of AMPK phosphorylation and regulation of den-
spine morphology have been shown in the dorsal dritic spines. APPL1 deficiency exacerbated the
spinal cord and were associated with indices of hyperalgesia in STZ-rats, and restoration of
neuropathic pain [61]. Morphologic changes APPL1 levels ameliorated painful diabetic neu-
were not observed after 1 week of diabetes when ropathy [72].
hyperglycemia was present without evidence of Sirtuins also play a role in synaptic plasticity
pain [61]. Additionally, evoked responses to and pain. Sirtuin levels were shown to be
brush and low force von Frey filaments, but not decreased along with enhanced expression of
higher force static von Frey stimulation, were structural synaptic plasticity protein (PSD95,
recorded from wide dynamic range (WDR) neu- GAP 43 and synaptophysin) in spinal dorsal horn
rons in STZ diabetic rat spinal cord while, in the of diabetic neuropathic rats and db/db mice [73,
genetic BB/Wor rat model, responses to brush 74]. Activator of Sirtuin 1 relieved pain behavior
and von Frey stimulation were not affected, cor- and inhibited the enhanced structural synaptic
responding to their lack of tactile hypersensitivity plasticity in diabetic rats and db/db mice while
[62]. ­silencing Sirtuin 1-induced pain behavior in nor-
In neuronal cells, RhoA, a member of the mal rats [73].
small molecular G proteins family, Ras super- Preclinical work in animal models of diabetes
family (see review [63]), is involved in the guid- demonstrated alteration of synaptic plasticity at
ance and extension of axons as well as the the spinal cord levels contributing to painful dia-
368 A. G. Marshall et al.

betic neuropathy, which corroborated with human intrathecal MK801 reduced mechanical allodynia
data, where MRI imaging showed spinal cord along with reduction of pERK, pAKT, TNFα,
atrophy as an early process only in patients with and IL6 levels in the spinal cord [41].
clinically detectable diabetic painful neuropathy Synaptic calcium-permeable AMPA receptors
[4]. are increased in the spinal dorsal neurons of dia-
betic rats and blocking them reduced nociceptive
hypersensitivity [81]. Increased metabotropic
3.3 Receptors and Ion Channels glutamate receptors (mGluR5) in spinal cord of
STZ diabetic rats may also contribute to increased
Pain, and more particularly here, diabetic painful glutamatergic input and nociceptive transmission
neuropathy, is linked to aberrant, exaggerated, or in diabetic neuropathic pain [82].
reduced receptors and ion channels activity.
Preclinical models of painful diabetic neuropathy 3.3.2 Adrenergic Receptors
have allowed studies of the pathological modula- Adrenergic receptors are G-protein-coupled
tion of ion channels at the spinal cord level and receptors, with two main groups, α and β, in
the development of therapies. which activation can generate analgesic effect or
contribute to chronic neuropathic pain [83]. The
3.3.1 Glutamatergic Receptors increased amplitude of glutamatergic excitatory
mRNA for subunits of glutamatergic NMDA and postsynaptic current in spinal cord slides from
AMPA receptors are increased in the spinal cord STZ diabetic rats was inhibited by a specific
of diabetic rodents [75]. More specifically, α2-adrenoceptor agonist, which concurrently
NMDA R1 subunit levels were increased in the alleviated hyperalgesia. The electrophysiological
spinal dorsal horn of STZ diabetic rats which study suggested an upregulation of
were hyperalgesic/allodynic, and magnesium, a α2-adrenoceptors activity in the spinal cord horn
voltage-dependent blocker of NMDA, prevented of STZ diabetic rats and a somewhat contrastive
the increased of phosphorylated NR1 (activation) antinociceptive effect of the α2 adrenoceptor
and thermal and tactile allodynia [16, 76]. Similar agonist that resulted in attenuation of glutamater-
results were obtained with the NMDA antagonist gic transmission in the spinal dorsal horn [84]. In
MK-801 [77]. Additionally, higher expression of allodynic db/db mice, decreased levels of neuro-
phosphorylated spinal NR2B subunit was accom- nal α2 adrenergic receptor protein were detected
panied by tactile allodynia and increased noci- during the period of mechanical allodynia, which
ceptive response in the formalin test in diabetic was alleviated by systemic administration of dex-
rats and was reduced by intrathecal treatment medetomidine, a selective α2 adrenergic receptor
with ifenprodil, an atypical non-competitive agonist [85], possibly via the Wnt10a/β catenin
NMDA R2B antagonist [78]. Expression of spi- signaling pathway [33].
nal NMDA R2B was reduced along with TRPV1
expression in diabetic mice after treatment with 3.3.3 Ligand-Gated Channels
Ginger extracts and was associated with partial Transient receptor potential (TRP) channels
alleviation of allodynia and thermal hyperalgesia transduce extracellular stimuli into neuronal
[79]. Activation of NMDA R was also demon- responses through influx of calcium [86] and
strated in prediabetes and type 2 diabetes. Spinal when dysfunctional may contribute to painful
NMDA R2 expression and activation were diabetic neuropathy. Protein expression of TRP
increased in high-fat-diet-fed mice at 24 weeks vanilloid 1 (TRPV1) was increased at an early
when tactile allodynia and thermal hyperalgesia stage in the spinal cord, but not the DRG, of STZ
occurred [80]. Furthermore, inhibition of NMDA rats developing allodynia. The increased TRPV1
R2B alleviated tactile allodynia but not thermal levels were detected in small size CGRP neurons
hypoalgesia in the prediabetic mice [80]. In type and intrathecal administration of TRPV1 antago-
2 diabetic mice (db/db), NMDA inhibition by nists alleviated allodynia [87]. Similarly,
The Spinal Cord in Diabetic Neuropathy 369

increased expression of TRPV1 receptor in the responses with intrathecal administrations of


spinal cord of STZ diabetic rats was reduced by T-channel blockers rapidly reducing neural excit-
treatment with the antidepressant mirtazapine ability and pain responses [95, 96]. Only one
[88]. Recently, in contrast, TRPV1 and CGRP study has demonstrated that specific inhibition of
expressions were shown to be reduced in the spi- Cav3.2 T channels in superficial dorsal horn neu-
nal cord of diabetic rats and further decreased rons suppressed spontaneous excitatory synaptic
with administration of Ropivacaine, a commonly transmission in diabetic rats to a greater extent
used clinical anesthetic, dramatically affecting than in healthy age-matched animals [97].
NCV of the sciatic nerve [89], suggesting greater Aberrant regulation of nociceptive receptors
precaution when planning surgery for patients and ion channels in the dorsal horn of diabetic
with diabetes. rats contribute to increased pronociception via
Spinal TRP ankyrin 1 (A1) channels, on cen- ion channels in sensory neurons. Modulation of
tral terminals of primary afferent nerve fibers, spinal receptors and channels may lead to novel
were shown to play an important role in mainte- therapies for painful diabetic neuropathy.
nance of mechanical hypersensitivity and con-
tribute to cutaneous neurogenic inflammation
while cutaneous TRPA1 channels contributed to 3.4 Oxidative Stress
mechanical hypersensitivity in STZ rats [90, 91].
Methylglyoxal, a reactive glucose metabolite, Reactive oxygen species (ROS) are produced by
was reported to facilitate painful diabetic neu- physiological functions and scavenged by enzy-
ropathy via sensitization of TRPA1-adenylyl matic and non-enzymatic antioxidant system. In
cyclase type 1 (AC1) signaling cascade in type 2 diabetes, chronic hyperglycemia leads to an
diabetic db/db mice [92]. imbalance of the oxidative status [98] and
increased oxidative stress occurs in peripheral
3.3.4 Voltage-Gated Sodium and central tissues [99]. Several studies have
Channels demonstrated the role that oxidative stress plays
Voltage-gated sodium channels (Nav) play a criti- at the level of the spinal cord to contribute to dia-
cal role in controlling cellular excitability and betic neuropathic pain, mainly by demonstrating
have gain interest for their role in painful diabetic the beneficial effect of antioxidants.
neuropathy particularly Nav 1.3, 1.7, and 1.8 Poly(ADP-ribose)Polymerase-1 (PARP) is an
among the 10 sodium channel α subunits nuclear enzyme that cleaves nicotinamide ade-
(Nav1.1-­1.9 and NavX). nine dinucleotide (NAD+) and contributes to
Intrathecal administration of Nav1.7 and NAD+ deletion and energy failure, impaired sig-
Nav1.8 antagonists alleviated thermal hyperalge- nal transduction, and apoptosis [100]. Enhanced
sia in diabetic mice but not in control mice, activation of PARP was demonstrated in diabetic
although changes in the spinal sodium channels rat spinal cord, with increased nitrotyrosine lev-
proteins were not different between control and els in neurons, oligodendrocytes, and astrocytes.
diabetic mice [93]. A PARP inhibitor prevented sciatic nerve and
spinal PARP activation, nitrotyrosine accumula-
3.3.5 Voltage-Gated Calcium tion, and ameliorated the neuropathy in the STZ
Channels rats [48].
Voltage-gated calcium channels of L, N, and T Curcumin is a natural polyphenol with multi-
type are widely studied for their role in neuro- ple properties, the most recognized being
pathic pain, with the T-type channel Cav3.2 par- antioxidant and anti-inflammatory [101].
­
ticularly studied in painful diabetic neuropathy, Curcumin was shown to ameliorate diabetic neu-
mainly in DRG sensory neurons [94]. Limited ropathy by suppression of oxidative stress in
numbers of studies have indirectly shown a role brain and sciatic nerves of diabetic rats [102] and
of dorsal horn T-channels in neuropathic pain recently was shown to reduce oxidative stress
370 A. G. Marshall et al.

markers in the spinal cord of STZ diabetic rats sion of TLR4-MYd88-dependent pathway [111].
via inhibition of the spinal NADPH oxidases Other antidepressants, milnacipan (SNRI), par-
[103]. Consistently with its anti-inflammatory oxetine and fluvoxamine (SSRI) [112], and
properties, curcumin was shown to reduce ammoxetine (SNRI) [32] alleviated allodynia in
expression of TNFα and TNFα receptor 1 in the STZ diabetic rats after intrathecal administration.
dorsal horn of STZ diabetic rats [27]. Despite the influence of serotonin in pain modu-
Increased oxidative stress damage and neuro- lation, SSRIs are less effective than tricyclic anti-
nal activation (Fos) in the spinal cord neurons depressants. 5HT2A receptor density was not
were normalized by 10 weeks of preventative affected in the spinal cord of diabetic rats [109,
treatment of STZ rats with an antioxidant 113], but the PDZ domains of PSD-95 that inter-
(epigallocatechin-­gallate) [104], by mexiletine, a acts with 5HT2A receptors was upregulated. This
sodium channel blocker with antioxidative prop- interaction PDZ/5HT2AR might contribute to the
erties [105] and by treatment with ozone and/or resistance of SSRI-induced analgesia in painful
insulin [106]. diabetic neuropathy, as disrupting this interaction
Increased oxidative stress has been demon- enhanced fluoxetine antihyperalgesic effect
strated in diabetic neuropathy, and the ability to [113].
scavenge ROS ameliorates diabetic neuropathy Aberrant regulation of nociceptive receptors
in rodents. and ion channels in the dorsal horn of diabetic
rats contributes to increased pronociception via
ion channels in sensory neurons. Modulation of
3.5 Serotonin/Noradrenaline spinal receptors and channels may lead to novel
Reuptake Inhibitors therapies for painful diabetic neuropathy.

Among the four currently FDA-approved drugs


to improve painful diabetic polyneuropathy 4 Spinal Disinhibition
[107], two are serotonin/noradrenaline reuptake
inhibitors (SSRI/SNRI), duloxetine and fluox- Depending on the physiological and pathophysi-
etine, although fluoxetine is supported by only a ological state, processing in the spinal cord dor-
small study. Duloxetine was the first SSRI pre- sal horn may dampen down or enhance output
scription drug approved by FDA for the manage- from nociceptive projection neurons. Several
ment of pain associated with diabetic neuropathy possible mechanisms of spinally mediated
[108] and is the most recommended and pre- enhancement of ascending nociceptive drive
scribed treatment for painful diabetic neuropathy including wind-up, long-term potentiation, glial
while studies continued to define its mechanism(s) inflammation, altered descending pain modula-
of action. Duloxetine exerted its anti-allodynic tion, and spinal disinhibition have been impli-
effect in diabetic rats predominantly via indirect cated in centrally mediated pain. The following
activation of 5HT2A receptors in the spinal cord section will focus on the accumulating evidence
[109]. Duloxetine is a balanced serotonin/nor- suggesting that spinal disinhibition plays an
adrenaline reuptake inhibitor, and its efficacy important role in pain generation in diabetic
may also depend on modulation of noradrenaline, neuropathy.
as it reduced noradrenergic signals in the spinal The major inhibitor transmitters in the spinal
cord by inhibiting the norepinephrine transport- cord are γ-aminobutyric acid (GABA) and gly-
ers [110]. Other mechanisms may play a role in cine. GABA acts via GABA-A and GABA-B to
the analgesic effect of duloxetine. In diabetic produce inhibition either at pre-synaptic termi-
mice, duloxetine treatment for 4 weeks alleviated nals, thereby reducing transmitter release (i.e.,
allodynia and thermal hyperalgesia, and this was pre-synaptic inhibition) or on the post-synaptic
associated with reduction of microglia and astro- dendrites (i.e., post-synaptic inhibition). GABA-­
cytes expression [38] and reduced overexpres- A-­mediated inhibition is ionotropic resulting
The Spinal Cord in Diabetic Neuropathy 371

from the passage of ions through voltage-gated ioral indices of pain in rodent pain models
channels [114]. The main ion that permeates the [128–131]. A reduction in this tonic GABAergic/
pore of the GABA-A receptor is chloride (Cl−), glycinergic inhibition of ascending nociceptive
the concentration of which is much greater in the transmission, a phenomenon termed disinhibi-
extracellular space. The polarity of the GABA-A tion, is a putative mechanism of central sensitiza-
response is determined largely by the intracellu- tion and could potentially result from loss of
lar Cl− concentration. In the post-developmental inhibitory interneurons or attenuation of inhibi-
nervous system, the low intracellular Cl− concen- tory neurotransmitter storage/release [132, 133].
tration is controlled by the potassium (K+)/chlo- Indeed, a reduction in GABAergic tone in the
ride (Cl−) co-transporter KCC2 which extrudes dorsal horn of the spinal cord has been demon-
intracellular Cl− [115]. Following GABA-A acti- strated in models of peripheral nerve injury [134,
vation, Cl− enters the cell according to its electro- 135]. However, this is not the case in the spinal
chemical concentration gradient, causing cord of diabetic rats exhibiting behavioral indices
inhibitory post-synaptic potentials (IPSP), lead- of pain. In the STZ rat model of type 1 diabetes,
ing to phasic hyperpolarization [114]. It is concentrations of GABA in the spinal cord are
increasingly recognized that subtypes of increased in both basal state and evoked responses
GABA-A also occur at sites other than the syn- in the formalin-induced pain model. In contrast,
apse. These extrasynaptic GABA-A receptors, in the same study, levels of the excitatory neu-
which are activated by GABA derived from glial rotransmitter glutamate are diminished [136].
cells and “circulating” GABA [116], produce Given that spinal cord GABA-A receptor protein
tonic hyperpolarization and consequently tonic levels are not significantly altered in STZ rats
inhibition of spinal cord cells [117, 118] and pri- [137], the paradoxical co-existence of increased
mary afferents [119]. GABA and increased nociceptive drive suggest
GABA-B-mediated inhibition is, in contrast, that there is impaired/altered post-synaptic
metabotropic. The GABA-B receptor is G-protein responsiveness to GABA.
coupled, and the binding of GABA triggers a sec- Alterations in the post-synaptic responsive-
ond messenger cascade [120, 121]. In the post-­ ness to GABA were first described in a peripheral
synaptic cell, this results in activation of nerve injury model of pain in nondiabetic rats
G-protein-activated inwardly rectifying K+ [138]. Increasing evidence indicates that the dys-
(GIRK) channels which are highly co-localized functional spinal inhibitory processing seen in
with GABA-B receptors [120–122]. The result- STZ rats results from a similar mechanism.
ing increase in K+ conductance promotes K+ Allodynia and other behavioral indices of pain in
efflux from the cell and hyperpolarization. In STZ rats are associated with downregulation of
contrast to the fast inhibitory signaling mediated post-synaptic KCC2  in the dorsal, although not
by GABA-A receptors, GABA-B receptor-­ ventral, spinal cord [137, 139]. This causes
mediated hyperpolarization is slow and pro- GABA, acting via spinal GABA-A receptors, to
longed [121]. become pro-nociceptive rather than inhibitory.
Inhibitory interneurons in the spinal cord are Accordingly, spinal GABA-A blockade, which
intimately associated with nociceptive circuits under normal circumstances is pro-nociceptive,
[123]. The effect of this inhibition is to reduce the reverses allodynia in STZ rats [137]. The mecha-
output of projection neurons in response to nox- nism, like that seen in the peripheral nerve injury
ious peripheral stimulation. Accordingly, in nor- model, also involves BDNF.  Spinal administra-
mal rodents, spinal application of GABA or tion of BDNF in normal rats phenocopies the
glycine antagonists result in behavioral indices of findings in STZ rats with the emergence of allo-
hypersensitivity and pain to both noxious and dynia and reduced spinal KCC2 expression
innocuous tactile stimulation [124–127]. [139]. Furthermore, sequestration of spinal
Conversely, enhancement of spinal GABA ele- BDNF in STZ rats reverses allodynia [139]. The
vates nociceptive thresholds and reverses behav- source of the BDNF in STZ rats is currently not
372 A. G. Marshall et al.

clear, although, interestingly, BDNF expression on synaptic GABA-A.  For example, altered
is elevated in the dorsal root ganglia of STZ rats, responsiveness of the α5 subunit-containing
raising the possibility that it may derive from the GABA-A receptor is also implicated in allo-
primary afferent fibers [140]. dynia and impaired RDD in STZ rats [153].
Inhibitory function in the spinal cord can be Normal levels of RDD can also occur in the
assessed using rate-dependent depression (RDD) presence of GABA-A blockade. As we have
of the Hoffmann-reflex (H-reflex) [141]. The already discussed, spinal disinhibition and
H-reflex is a trans-spinal reflex that is elicited by impaired RDD are reversed by GABA-A block-
electrical stimulation of a mixed motor and sen- ing drugs in normal rats treated with KCC2
sory nerve trunk. The H-reflex can be measured antagonist DOIA or BDNF as well as in diabetic
in mammalian species including rodents and rats [137]. This implies that other inhibitory
humans using a simple modification of traditional mechanisms can also contribute to RDD in the
nerve conduction studies [142–145]. When a sec- spinal cord of STZ rats. Further evidence for
ond H-wave is elicited with an inter-stimulus this is revealed in studies investigating the time
interval less than 2 s following the initial stimula- course of RDD and spinal disinhibition in STZ
tion, the H-wave amplitude decreases relative to rats which indicate that the development of spi-
the first response. The measure of the change in nal disinhibition in diabetes is a dynamic time-
amplitude is referred to as H-reflex RDD—also dependent process involving GABA-A- and
known as paired-pulse depression or frequency-­ GABA-B-mediated responses [152]. Impaired
dependent depression [146–151]. RDD due to GABA-A reversal is initially com-
The plausibility of RDD as a mechanistic pensated by GABA-B mediated inhibition.
biomarker of loss of GABAergic inhibition due However, over time, due to increasing GABA-A
to GABA reversal, spinal disinhibition, and reversal or attenuation of GABA-B inhibitory
resultant pain-related phenomena in painful function, impaired RDD and the full spinal dis-
diabetic neuropathy has been demonstrated in a inhibition phenotype become evident. The role
number of studies in rodents. STZ rats exhibit- of glycinergic inhibition in spinal disinhibition
ing mechanical hypersensitivity/allodynia and in diabetic rats has not been studied extensively
reduced KCC2 in the dorsal horn show attenua- although glycine release in the spinal cord of
tion of RDD [137]. Furthermore, spinal deliv- STZ rats are mildly reduced [136].
ery of either the KCC2 blocker DIOA or BDNF The disclosure of spinal disinhibition as a
to control rats, interventions that mechanisti- putative pain mechanism in STZ rats raises the
cally recapitulate the spinal disinhibition phe- question as to whether similar mechanisms occur
notype in STZ rats, are also associated with in clinical populations with painful diabetic neu-
impaired RDD [137, 139]. In keeping with the ropathy. This is of potential clinical importance
underlying mechanism of GABA reversal, both as it may lead to mechanistically targeted drug
the behavioral indices of pain and loss of RDD discovery or individualized therapy in patient
in all these models are reversed by spinal populations. However, this would require identi-
administration of GABA-A antagonists fication of populations or sub-populations of
[137]—a pharmacological intervention that patients with painful diabetic neuropathy in
both induces hypersensitivity and impairs RDD whom spinal disinhibition is a major pain mecha-
in control rats. RDD and allodynia in STZ rats nism. Currently it is not possible to measure lev-
are also reversed by the carbonic anhydrase els of dorsal horn GABA or KCC2  in humans.
inhibitor acetazolamide, which mitigates the However, RDD is conserved across mammalian
effects of reduced KCC2 and Cl− dysregulation species including humans raising the possibility
by decreasing bicarbonate efflux through that results obtained in rodents may be translated
GABA-A receptors [152]. to diabetic patients.
Spinal disinhibition and loss of RDD in pain- To assess the translational potential of the
ful diabetic neuropathy are not just dependent preclinical findings, we measured the magni-
The Spinal Cord in Diabetic Neuropathy 373

tude of H-reflex RDD in patients with type 1 receptor function. Although the exact neural cir-
diabetes and painful or painless neuropathy cuitry has yet to be defined, both RDD and
[154]. In a subset of patients with painful dia- behavioral indices of neuropathic pain also
betic neuropathy, there was loss of RDD com- exhibit common responses to spinally acting
pared to both healthy controls and patients with analgesics. RDD status, which is easily measur-
painless diabetic neuropathy. Importantly, the able in clinical populations, may be a viable bio-
impairment of RDD was independent of mea- marker for identifying the dominant generator
sures of both large and small fiber neuropathy, site in individual patients with painful diabetic
indicating that is not merely a reflection of neuropathy and for predicting efficacy of thera-
severity of neuropathy. Loss of RDD was also peutic strategies that alleviate spinal
independent of glycemic control. These findings disinhibition.
support the hypothesis that impaired RDD may
serve as clinical biomarker in a subset of patients
where pain arises primarily from spinal disinhi- 5 Spinal Cord Stimulation
bition. We have also recently demonstrated that
impaired RDD is also seen in patients with type Spinal cord stimulation (SCS) is proposed to
2 diabetes and painful neuropathy (unpublished relieve chronic intractable pain by stimulating
observations). Like the findings in type 1 diabe- nerve fibers in the spinal cord. The resulting
tes, not all subjects with type 2 diabetes and impulses in the fibers may inhibit the conduction
neuropathic pain demonstrated impairment of of pain signals to the brain, according to the pain
RDD. Approximately 60% of patients with dia- gate theory proposed by Melzack and Wall [156]
betes will develop neuropathy, 30% of those and block the sensation of pain. In general, SCS
with neuropathy will develop neuropathic pain is part of an overall treatment strategy and is used
and, from our exploratory study, 40% of those only after the more conservative treatments have
will show RDD deficits. In contrast, diabetic failed (review in [157]). SCS is an effective ther-
rodents exhibit much more homogeneous neu- apy for different chronic painful conditions that
ropathy, neuropathic pain, and impaired RDD has gained interest for painful peripheral diabetic
phenotypes [141]. While this may reflect a more neuropathy within the last 15 years. In addition to
complex etiopathogenesis of painful diabetic the pain gate theory, several other mechanisms
neuropathy in humans, it is also plausible that have been proposed for the beneficial effect of
this heterogeneity can be used to enable defini- SCS in diabetes (review in [158]). SCS may pro-
tion of abnormal values and predict therapeutic duce peripheral vasodilatation via stimulation of
response to medications that target spinal inhi- the sympathetic system and release of neuro-
bition, one of these drugs being duloxetine. The trophic factors [159, 160] and may involve small
selective serotonin-norepinephrine re-uptake diameter nociceptive afferents expressing TRPV1
inhibitor duloxetine, used to treat painful dia- receptor which stimulation induces release of
betic neuropathy [155], alleviates the tactile calcitonin gene-related peptide (CGRP), nitric
allodynia and also restores RDD in STZ rats in oxide [161] and activation of ERK [162].
a spinal 5HT2A receptor-dependent manner Vasodilatation was observed after SCS in STZ
[109, 154], and diabetic patients with impaired diabetic rats; however, hyperglycemic conditions
spinal disinhibition seemed to find duloxetine to attenuated the blood flow responses to SCS [163],
be an efficacious treatment for their neuropathic possibly due to the decreased CGRP levels [164,
pain [154]. 165], or reduced TRPV1 containing C fibers
To summarize, there is accumulating preclini- [161]. Another mechanism may involve altera-
cal evidence that loss of RDD and indices of neu- tion of spinal neurochemistry, such as increased
ropathic pain share a common pathogenic GABA and acetylcholine after SCS resulting in
mechanism involving spinal KCC2 depletion and the activation of inhibitory interneurons with the
disinhibition caused by inversion of GABA-A reduction of allodynia in neuropathic pain mod-
374 A. G. Marshall et al.

els [166, 167]. This plausible mechanism has not 6 Summary/Conclusions


been studied in diabetic models and may not
apply to efficacy of SCS in diabetes. GABA has It is clear, particularly from the preclinical
been shown to be excitatory in diabetic rats due rodent studies, that diabetes is a multifactorial
to impairment in spinal KCC2, resulting in spinal disease. Many mechanisms take place in the
inhibitory dysfunction (see Sect. 4 and [136, 137, spinal cord, contributing to neuropathy and
154]). painful neuropathy that can be alleviated in ani-
Mechanisms by which SCS improves neuro- mal models by a multitude of drugs. However,
pathic pain, particularly in diabetes, are not little has been translated to human with many
clearly understood. However, studies in diabetic failed clinical trials. Nevertheless, progress has
patients have shown efficacy. Neuropathic pain been made with spinal cord stimulation to ben-
was significantly reduced after 6 months of SCS efit intractable pain, four FDA-approved drugs
in diabetic patients with chronic pain in their are now available, and a new method of detec-
lower limbs that was not responsive to conven- tion of pain generation site may facilitate physi-
tional treatment, and this was not associated with cians’ prescription ability of the current drugs.
changes in microcirculatory perfusion [168]. The detection of pain generation site by RDD
SCS showed pain relief of more than 50% in may also enhance clinical trial design and
50–77% of patients [169, 170] and up to 5 years patient selection for central versus peripheral
[171]. effectors and therefore contribute to more suc-
Optimization of SCS parameters has shown cessful clinical trials.
improvement over conventional SCS (30–
120  Hz). One of these optimization is high-­
frequency SCS (150–500  Hz). In a preclinical References
rodent model of type 1 diabetes, alleviation of
mechanical hypersensitivity was independent of 1. Mizisin AP, Jolivalt CG, Calcutt NA (2007) Spinal
cord: structure and function in diabetes. In: Veves A,
the frequency of stimulation applied [172]. Malik R (eds) Contemporary diabetes: diabetic neu-
However, it was shown that high-frequency SCS ropathy: clinical management, 2nd edn
demonstrated delayed effect after cessation of 2. Calcutt NA, Freshwater JD, O'Brien JS.  Protection
SCS on mechanical allodynia in STZ diabetic of sensory function and antihyperalgesic proper-
ties of a prosaposin-derived peptide in diabetic rats.
rats [173]. Anesthesiology. 2000;93:1271–8.
Recently, another paradigm of stimulation has 3. Eaton SE, Harris ND, Rajbhandari SM, et al. Spinal-­
been developed, burst SCS (5 pulses at 500 Hz, cord involvement in diabetic peripheral neuropathy.
delivered 40  times/s). Burst SCS reduced pain Lancet. 2001;358:35–6.
4. Selvarajah D, Wilkinson ID, Emery CJ, et al. Early
further than tonic stimulation (30–120 Hz) with involvement of the spinal cord in diabetic peripheral
reduced local paresthesia [174]. About 58% of neuropathy. Diabetes Care. 2006;29:2664–9.
the patients experienced significant additional 5. Tesfaye S, Selvarajah D, Gandhi R, et  al. Diabetic
pain reduction [175]. peripheral neuropathy may not be as its name sug-
gests: evidence from magnetic resonance imaging.
SCS has improved in the last 15 years with Pain. 2016;157(Suppl 1):S72–80.
high frequency and burst protocols applied to 6. Quan Y, Du J, Wang X.  High glucose stimu-
diabetes-induced neuropathic pain not respond- lates GRO secretion from rat microglia via ROS,
ing to current treatments. However, little is PKC, and NF-kappaB pathways. J Neurosci Res.
2007;85:3150–9.
known in diabetic conditions and the mecha- 7. Quan Y, Jiang CT, Xue B, Zhu SG, Wang X. High
nisms by which SCS is effective in diabetic glucose stimulates TNFalpha and MCP-1 expression
patients remain unknown. Further studies are in rat microglia via ROS and NF-kappaB pathways.
needed to understand how SCS works in diabetes Acta Pharmacol Sin. 2011;32:188–93.
8. Daulhac L, Mallet C, Courteix C, et  al. Diabetes-­
and to predict which patients may benefit from induced mechanical hyperalgesia involves
SCS, possibly patients with evidence of spinal spinal mitogen-activated protein kinase activa-
disinhibition detected by RDD. tion in neurons and microglia via N-methyl-D-­
The Spinal Cord in Diabetic Neuropathy 375

aspartate-dependent mechanisms. Mol Pharmacol. protein expression in rat spinal cord. J Diabetes
2006;70:1246–54. Metab Disord. 2019;18:181–90.
9. Cheng KI, Wang HC, Chuang YT, et  al. Persistent 21. Ismail CAN, Suppian R, Ab Aziz CB, Long
mechanical allodynia positively correlates with I. Expressions of spinal microglia activation, BDNF,
an increase in activated microglia and increased and DREAM proteins correlated with formalin-­
P-p38 mitogen-activated protein kinase activation induced nociceptive responses in painful and
in streptozotocin-induced diabetic rats. Eur J Pain. painless diabetic neuropathy rats. Neuropeptides.
2014;18:162–73. 2020;79:102003.
10. Toth CC, Jedrzejewski NM, Ellis CL, Frey WH 2nd. 22. Sun JJ, Tang L, Zhao XP, Xu JM, Xiao Y, Li
Cannabinoid-mediated modulation of neuropathic H.  Infiltration of blood-derived macrophages con-
pain and microglial accumulation in a model of tributes to the development of diabetic neuropathy.
murine type I diabetic peripheral neuropathic pain. J Immunol Res. 2019;2019:7597382.
Mol Pain. 2010;6:16. 23. Afsari ZH, Renno WM, Abd-El-Basset E. Alteration
11. Suzuki N, Hasegawa-Moriyama M, Takahashi Y, of glial fibrillary acidic proteins immunoreactivity in
Kamikubo Y, Sakurai T, Inada E. Lidocaine attenu- astrocytes of the spinal cord diabetic rats. Anat Rec
ates the development of diabetic-induced tactile (Hoboken). 2008;291:390–9.
allodynia by inhibiting microglial activation. Anesth 24. Renno WM, Alkhalaf M, Afsari Z, Abd-El-Basset
Analg. 2011;113:941–6. E, Mousa A.  Consumption of green tea alters glial
12. Thakur V, Gonzalez M, Pennington K, Nargis S, fibriliary acidic protein immunoreactivity in the
Chattopadhyay M. Effect of exercise on neurogenic spinal cord astrocytes of STZ-diabetic rats. Nutr
inflammation in spinal cord of type 1 diabetic rats. Neurosci. 2008;11:32–40.
Brain Res. 2016;1642:87–94. 25. Kim SH, Kwon JK, Kwon YB.  Pain modality and
13. Wodarski R, Clark AK, Grist J, Marchand F, spinal glia expression by streptozotocin induced dia-
Malcangio M. Gabapentin reverses microglial acti- betic peripheral neuropathy in rats. Lab Anim Res.
vation in the spinal cord of streptozotocin-induced 2012;28:131–6.
diabetic rats. Eur J Pain. 2009;13:807–11. 26. Byrne FM, Cheetham S, Vickers S, Chapman
14. Morgado C, Pereira-Terra P, Cruz CD, Tavares V. Characterisation of pain responses in the high fat
I.  Minocycline completely reverses mechanical diet/streptozotocin model of diabetes and the analge-
hyperalgesia in diabetic rats through microglia-­ sic effects of antidiabetic treatments. J Diabetes Res.
induced changes in the expression of the potassium 2015;2015:752481.
chloride co-transporter 2 (KCC2) at the spinal cord. 27. Li Y, Zhang Y, Liu DB, Liu HY, Hou WG, Dong
Diabetes Obes Metab. 2011;13:150–9. YS.  Curcumin attenuates diabetic neuropathic pain
15. Tsuda M, Ueno H, Kataoka A, Tozaki-Saitoh H, by downregulating TNF-alpha in a rat model. Int J
Inoue K.  Activation of dorsal horn microglia con- Med Sci. 2013;10:377–81.
tributes to diabetes-induced tactile allodynia via 28. Liu M, Liao K, Yu C, Li X, Liu S, Yang S. Puerarin
extracellular signal-regulated protein kinase signal- alleviates neuropathic pain by inhibiting neuro-
ing. Glia. 2008;56:378–86. inflammation in spinal cord. Mediators Inflamm.
16. Daulhac L, Maffre V, Mallet C, et al. Phosphorylation 2014;2014:485927.
of spinal N-methyl-d-aspartate receptor NR1 sub- 29. Newton VL, Guck JD, Cotter MA, Cameron NE,
units by extracellular signal-regulated kinase in Gardiner NJ.  Neutrophils infiltrate the spinal cord
dorsal horn neurons and microglia contributes to parenchyma of rats with experimental diabetic neu-
diabetes-induced painful neuropathy. Eur J Pain. ropathy. J Diabetes Res. 2017;2017:4729284.
2011;15:169.e161–12. 30. Yao L, Wu YT, Tian GX, Xia CQ, Zhang F, Zhang
17. Johnson MS, Ryals JM, Wright DE.  Diabetes-­ W.  Acrolein scavenger hydralazine prevents
induced chemogenic hypoalgesia is paralleled by streptozotocin-­ induced painful diabetic neuropa-
attenuated stimulus-induced fos expression in the thy and spinal neuroinflammation in rats. Anat Rec
spinal cord of diabetic mice. J Pain. 2007;8:637–49. (Hoboken). 2017;300:1858–64.
18. Morgado C, Terra PP, Tavares I.  Neuronal hyper- 31. Liu M, Qiang QH, Ling Q, et al. Effects of Danggui
activity at the spinal cord and periaqueductal grey Sini decoction on neuropathic pain: experimental
during painful diabetic neuropathy: effects of gaba- studies and clinical pharmacological significance of
pentin. Eur J Pain. 2010;14:693–9. inhibiting glial activation and proinflammatory cyto-
19. Pabreja K, Dua K, Sharma S, Padi SS, Kulkarni kines in the spinal cord. Int J Clin Pharmacol Ther.
SK. Minocycline attenuates the development of dia- 2017;55:453–64.
betic neuropathic pain: possible anti-inflammatory 32. Zhang TT, Xue R, Fan SY, et al. Ammoxetine atten-
and anti-oxidant mechanisms. Eur J Pharmacol. uates diabetic neuropathic pain through inhibiting
2011;661:15–21. microglial activation and neuroinflammation in the
20. Ismail CAN, Suppian R, Aziz CBA, Long spinal cord. J Neuroinflammation. 2018;15:176.
I.  Minocycline attenuates the development of dia- 33. Zhong JM, Lu YC, Zhang J.  Dexmedetomidine
betic neuropathy by modulating DREAM and BDNF reduces diabetic neuropathy pain in rats through the
376 A. G. Marshall et al.

Wnt 10a/beta-catenin signaling pathway. Biomed nerve growth factor deficit in diabetic mice. Eur J
Res Int. 2018;2018:9043628. Pharmacol. 2010;637:62–9.
34. Lee JY, Choi HY, Park CS, et al. GS-KG9 amelio- 47. Dogrul A, Gul H, Yesilyurt O, Ulas UH, Yildiz
rates diabetic neuropathic pain induced by strepto- O.  Systemic and spinal administration of etaner-
zotocin in rats. J Ginseng Res. 2019;43:58–67. cept, a tumor necrosis factor alpha inhibitor, blocks
35. Reda HM, Zaitone SA, Moustafa YM.  Effect of tactile allodynia in diabetic mice. Acta Diabetol.
levetiracetam versus gabapentin on peripheral neu- 2011;48:135–42.
ropathy and sciatic degeneration in streptozotocin-­ 48. Drel VR, Lupachyk S, Shevalye H, et al. New thera-
diabetic mice: influence on spinal microglia and peutic and biomarker discovery for peripheral dia-
astrocytes. Eur J Pharmacol. 2016;771:162–72. betic neuropathy: PARP inhibitor, nitrotyrosine,
36. Evangelista AF, Vannier-Santos MA, de Assis Silva and tumor necrosis factor-{alpha}. Endocrinology.
GS, et al. Bone marrow-derived mesenchymal stem/ 2010;151:2547–55.
stromal cells reverse the sensorial diabetic neuropa- 49. Liao YH, Zhang GH, Jia D, et al. Spinal astrocytic
thy via modulation of spinal neuroinflammatory cas- activation contributes to mechanical allodynia
cades. J Neuroinflammation. 2018;15:189. in a mouse model of type 2 diabetes. Brain Res.
37. Miranda HF, Poblete P, Sierralta F, Noriega V, 2011;1368:324–35.
Prieto JC, Zepeda RJ.  Interleukin-1beta in syner- 50. Dauch JR, Yanik BM, Hsieh W, Oh SS, Cheng
gism gabapentin with tramadol in murine model HT.  Neuron-astrocyte signaling network in spinal
of diabetic neuropathy. Inflammopharmacology. cord dorsal horn mediates painful neuropathy of type
2019;27:151–5. 2 diabetes. Glia. 2012;60:1301–15.
38. Tawfik MK, Helmy SA, Badran DI, Zaitone 51. Xu X, Chen H, Ling BY, Xu L, Cao H, Zhang
SA. Neuroprotective effect of duloxetine in a mouse YQ.  Extracellular signal-regulated protein kinase
model of diabetic neuropathy: role of glia suppress- activation in spinal cord contributes to pain hyper-
ing mechanisms. Life Sci. 2018;205:113–24. sensitivity in a mouse model of type 2 diabetes.
39. Elsherbiny NM, Ahmed E, Kader GA, et  al. Neurosci Bull. 2014;30:53–66.
Inhibitory effect of valproate sodium on pain behav- 52. Griggs RB, Donahue RR, Adkins BG, Anderson
ior in diabetic mice involves suppression of spinal KL, Thibault O, Taylor BK.  Pioglitazone inhibits
histone deacetylase 1 and inflammatory mediators. the development of hyperalgesia and sensitization of
Int Immunopharmacol. 2019;70:16–27. spinal nociresponsive neurons in type 2 diabetes. J
40. Ni GL, Cui R, Shao AM, Wu ZM. Salidroside ame- Pain. 2016;17:359–73.
liorates diabetic neuropathic pain in rats by inhibiting 53. Fernyhough P.  Mitochondrial dysfunction in dia-
neuroinflammation. J Mol Neurosci. 2017;63:9–16. betic neuropathy: a series of unfortunate metabolic
41. Liu S, Liu X, Xiong H, et  al. CXCL13/CXCR5 events. Curr Diab Rep. 2015;15:89.
signaling contributes to diabetes-induced tactile 54. Hamid HS, Mervak CM, Munch AE, et  al.
allodynia via activating pERK, pSTAT3, pAKT Hyperglycemia- and neuropathy-induced changes
pathways and pro-inflammatory cytokines produc- in mitochondria within sensory nerves. Ann Clin
tion in the spinal cord of male mice. Brain Behav Transl Neurol. 2014;1:799–812.
Immun. 2019;80:711–24. 55. Vincent AM, Edwards JL, McLean LL, et  al.
42. Leeb-Lundberg LM, Marceau F, Muller-Esterl W, Mitochondrial biogenesis and fission in axons in cell
Pettibone DJ, Zuraw BL.  International union of culture and animal models of diabetic neuropathy.
pharmacology. XLV.  Classification of the kinin Acta Neuropathol. 2010;120:477–89.
receptor family: from molecular mechanisms to 56. Chen L, Huang J, Li XC, et al. High-glucose induced
pathophysiological consequences. Pharmacol Rev. mitochondrial dynamics disorder of spinal cord
2005;57:27–77. neurons in diabetic rats and its effect on mitochon-
43. Regoli D, Nsa Allogho S, Rizzi A, Gobeil drial spatial distribution. Spine (Phila Pa 1976).
FJ. Bradykinin receptors and their antagonists. Eur 2019;44:E715–22.
J Pharmacol. 1998;348:1–10. 57. Rashedinia M, Alimohammadi M, Shalfroushan
44. Talbot S, Chahmi E, Dias JP, Couture R.  Key role N, et  al. Neuroprotective effect of syringic acid
for spinal dorsal horn microglial kinin B1 receptor in by modulation of oxidative stress and mito-
early diabetic pain neuropathy. J Neuroinflammation. chondrial mass in diabetic rats. Biomed Res Int.
2010;7:36. 2020;2020:8297984.
45. Ismail CAN, Aziz CBA, Suppian R, Long 58. Rashedinia M, Khoshnoud MJ, Fahlyan BK,
I. Imbalanced oxidative stress and pro-inflammatory Hashemi SS, Alimohammadi M, Sabahi Z. Syringic
markers differentiate the development of diabetic acid: a potential natural compound for the manage-
neuropathy variants in streptozotocin-induced dia- ment of renal oxidative stress and mitochondrial bio-
betic rats. J Diabetes Metab Disord. 2018;17:129–36. genesis in diabetic rats. Curr Drug Discov Technol.
46. Comelli F, Bettoni I, Colombo A, Fumagalli P, 2021;18(3):405–13.
Giagnoni G, Costa B.  Rimonabant, a cannabinoid 59. Malisza KL, Jones C, Gruwel ML, Foreman D,
CB1 receptor antagonist, attenuates mechani- Fernyhough P, Calcutt NA.  Functional magnetic
cal allodynia and counteracts oxidative stress and resonance imaging of the spinal cord during sensory
The Spinal Cord in Diabetic Neuropathy 377

stimulation in diabetic rats. J Magn Reson Imaging. mGluR1/5 expressions in type 2 diabetic rats. Pain.
2009;30:271–6. 2017;158:130–9.
60. Jiang Y, Mizisin AP, Rearden A, Jolivalt CG. 75. Tomiyama M, Furusawa K, Kamijo M, Kimura T,
Diabetes induces changes in ILK, PINCH and com- Matsunaga M, Baba M.  Upregulation of mRNAs
ponents of related pathways in the spinal cord of coding for AMPA and NMDA receptor subunits and
rats. Brain Res. 2010;1332:100–9. metabotropic glutamate receptors in the dorsal horn
61. Tan AM, Samad OA, Fischer TZ, Zhao P, Persson of the spinal cord in a rat model of diabetes mellitus.
AK, Waxman SG.  Maladaptive dendritic spine Brain Res Mol Brain Res. 2005;136:275–81.
remodeling contributes to diabetic neuropathic pain. 76. Rondon LJ, Privat AM, Daulhac L, et al. Magnesium
J Neurosci. 2012;32:6795–807. attenuates chronic hypersensitivity and spinal
62. Schuelert N, Gorodetskaya N, Just S, Doods H, cord NMDA receptor phosphorylation in a rat
Corradini L.  Electrophysiological characteriza- model of diabetic neuropathic pain. J Physiol.
tion of spinal neurons in different models of diabe- 2010;588:4205–15.
tes type 1- and type 2-induced neuropathy in rats. 77. Wang J, Sun Z, Wang Y, Wang H, Guo Y. The role
Neuroscience. 2015;291:146–54. and mechanism of glutamic NMDA receptor in the
63. Takai Y, Sasaki T, Matozaki T. Small GTP-binding mechanical hyperalgesia in diabetic rats. Neurol
proteins. Physiol Rev. 2001;81:153–208. Res. 2017;39:1006–13.
64. Nakayama AY, Harms MB, Luo L. Small GTPases 78. Ismail CAN, Suppian R, Abd Aziz CB, Haris K, Long
Rac and Rho in the maintenance of dendritic spines I. Increased nociceptive responses in streptozotocin-­
and branches in hippocampal pyramidal neurons. J induced diabetic rats and the related expression
Neurosci. 2000;20:5329–38. of spinal NR2B subunit of N-methyl-D-aspartate
65. Threadgill R, Bobb K, Ghosh A.  Regulation of receptors. Diabetes Metab J. 2019;43:222–35.
dendritic growth and remodeling by Rho, Rac, and 79. Fajrin FA, Nugroho AE, Nurrochmad A, Susilowati
Cdc42. Neuron. 1997;19:625–34. R.  Ginger extract and its compound, 6-shogaol,
66. Billuart P, Winter CG, Maresh A, Zhao X, Luo attenuates painful diabetic neuropathy in mice via
L. Regulating axon branch stability: the role of p190 reducing TRPV1 and NMDAR2B expressions in the
RhoGAP in repressing a retraction signaling path- spinal cord. J Ethnopharmacol. 2020;249:112396.
way. Cell. 2001;107:195–207. 80. Suo M, Wang P, Zhang M.  Role of Fyn-mediated
67. Ohsawa M, Kamei J. RhoA/Rho kinase signaling in NMDA receptor function in prediabetic neuropathy
the spinal cord and diabetic painful neuropathy. Eur in mice. J Neurophysiol. 2016;116:448–55.
J Pharmacol. 2010;644:1–4. 81. Chen SR, Zhang J, Chen H, Pan HL. Streptozotocin-­
68. Inoue M, Rashid MH, Fujita R, Contos JJ, Chun induced diabetic neuropathic pain is associated with
J, Ueda H.  Initiation of neuropathic pain requires potentiated calcium-permeable AMPA receptor
lysophosphatidic acid receptor signaling. Nat Med. activity in the spinal cord. J Pharmacol Exp Ther.
2004;10:712–8. 2019;371:242–9.
69. Tatsumi S, Mabuchi T, Katano T, et al. Involvement 82. Li JQ, Chen SR, Chen H, Cai YQ, Pan HL. Regulation
of Rho-kinase in inflammatory and neuropathic pain of increased glutamatergic input to spinal dorsal
through phosphorylation of myristoylated alanine-­ horn neurons by mGluR5  in diabetic neuropathic
rich C-kinase substrate (MARCKS). Neuroscience. pain. J Neurochem. 2010;112:162–72.
2005;131:491–8. 83. Carroll I, Gaeta R, Mackey S. Multivariate analysis
70. Mustafa S, Vasudevan H, Yuen VG, McNeill of chronic pain patients undergoing lidocaine infu-
JH. Renal expression of arachidonic acid metaboliz- sions: increasing pain severity and advancing age
ing enzymes and RhoA/Rho kinases in fructose insu- predict likelihood of clinically meaningful analge-
lin resistant hypertensive rats. Mol Cell Biochem. sia. Clin J Pain. 2007;23:702–6.
2010;333:203–9. 84. Chen SR, Chen H, Yuan WX, Pan HL.  Increased
71. Ohsawa M, Aasato M, Hayashi SS, Kamei J. RhoA/ presynaptic and postsynaptic alpha2-­ adrenoceptor
Rho kinase pathway contributes to the pathogen- activity in the spinal dorsal horn in pain-
esis of thermal hyperalgesia in diabetic mice. Pain. ful diabetic neuropathy. J Pharmacol Exp Ther.
2011;152:114–22. 2011;337:285–92.
72. He WY, Zhang B, Zhao WC, et  al. mTOR activa- 85. Chen H, Xu X, Yang XY, et  al. Systemic dexme-
tion due to APPL1 deficiency exacerbates hyperal- detomidine attenuates mechanical allodynia through
gesia via Rab5/Akt and AMPK signaling pathway extracellular sign db type 2 diabetic mice. Neurosci
in streptozocin-induced diabetic rats. Mol Pain. Lett. 2017;657:126–33.
2019;15:1744806919880643. 86. Julius D.  TRP channels and pain. Annu Rev Cell
73. Zhang Z, Ding X, Zhou Z, et  al. Sirtuin 1 allevi- Dev Biol. 2013;29:355–84.
ates diabetic neuropathic pain by regulating syn- 87. Cui YY, Xu H, Wu HH, Qi J, Shi J, Li YQ. Spatio-­
aptic plasticity of spinal dorsal horn neurons. Pain. temporal expression and functional involvement
2019;160:1082–92. of transient receptor potential vanilloid 1  in dia-
74. Zhou CH, Zhang MX, Zhou SS, et al. SIRT1 attenu- betic mechanical allodynia in rats. PLoS One.
ates neuropathic pain by epigenetic regulation of 2014;9:e102052.
378 A. G. Marshall et al.

88. Bektur E, Sahin E, Ceyhan E, et al. Beneficial effect 104. Raposo D, Morgado C, Pereira-Terra P, Tavares
of mirtazapine on diabetes-induced hyperalgesia: I.  Nociceptive spinal cord neurons of laminae I-III
involvement of TRPV1 and ASIC1 channels in the exhibit oxidative stress damage during diabetic
spinal cord and dorsal root ganglion. Neurol Res. neuropathy which is prevented by early antioxidant
2019;41:544–53. treatment with epigallocatechin-gallate (EGCG).
89. Zhang N, Wei H, Wu W, et al. Effect of ropivacaine Brain Res Bull. 2015;110:68–75.
on peripheral neuropathy in streptozocin diabetes-­ 105. Ates O, Cayli SR, Altinoz E, et al. Neuroprotective
induced rats through TRPV1-CGRP pathway. Biosci effect of mexiletine in the central nervous system of
Rep. 2019;39:BSR20190817. diabetic rats. Mol Cell Biochem. 2006;286:125–31.
90. Wei H, Chapman H, Saarnilehto M, Kuokkanen K, 106. Karademir M, Sonmez MA, Akcilar R, Kocak E,
Koivisto A, Pertovaara A. Roles of cutaneous versus Yay A, Eser O. Evaluation of therapeutic potential of
spinal TRPA1 channels in mechanical hypersensitiv- intraperitoneal ozone gas in combination with insulin
ity in the diabetic or mustard oil-treated non-diabetic above cranial and spinal neuropathy in rats with dia-
rat. Neuropharmacology. 2010;58:578–84. betes mellitus. Bratisl Lek Listy. 2018;119:636–41.
91. Wei H, Koivisto A, Pertovaara A. Spinal TRPA1 ion 107. Alam U, Sloan G, Tesfaye S.  Treating pain in dia-
channels contribute to cutaneous neurogenic inflam- betic neuropathy: current and developmental drugs.
mation in the rat. Neurosci Lett. 2010;479:253–6. Drugs. 2020;80:363–84.
92. Griggs RB, Santos DF, Laird DE, et al. Methylglyoxal 108. Lunn MP, Hughes RA, Wiffen PJ (2009) Duloxetine
and a spinal TRPA1-AC1-Epac cascade facilitate for treating painful neuropathy or chronic pain.
pain in the db/db mouse model of type 2 diabetes. Cochrane Database Syst Rev:CD007115.
Neurobiol Dis. 2019;127:76–86. 109. Mixcoatl-Zecuatl T, Jolivalt CG. A spinal mechanism
93. Tanaka K, Sekino S, Ikegami M, Ikeda H, Kamei of action for duloxetine in a rat model of painful dia-
J.  Antihyperalgesic effects of ProTx-II, a Nav1.7 betic neuropathy. Br J Pharmacol. 2011;164:159–69.
antagonist, and A803467, a Nav1.8 antagonist, in 110. Kinoshita J, Takahashi Y, Watabe AM, Utsunomiya
diabetic mice. J Exp Pharmacol. 2015;7:11–6. K, Kato F.  Impaired noradrenaline homeostasis in
94. Calcutt NA.  Diabetic neuropathy and neuropathic rats with painful diabetic neuropathy as a target of
pain: a (con)fusion of pathogenic mechanisms? Pain. duloxetine analgesia. Mol Pain. 2013;9:59.
2020;161:S65–86. 111. Zhou DM, Zhuang Y, Chen WJ, Li W, Miao
95. Maeda Y, Aoki Y, Sekiguchi F, et  al. Hyperalgesia B.  Effects of duloxetine on the toll-like recep-
induced by spinal and peripheral hydrogen sulfide: tor 4 signaling pathway in spinal dorsal horn in a
evidence for involvement of Cav3.2 T-type calcium rat model of diabetic neuropathic pain. Pain Med.
channels. Pain. 2009;142:127–32. 2018;19:580–8.
96. Matthews EA, Dickenson AH.  Effects of ethosuxi- 112. Ikeda T, Ishida Y, Naono R, et  al. Effects of intra-
mide, a T-type Ca(2+) channel blocker, on dorsal thecal administration of newer antidepressants on
horn neuronal responses in rats. Eur J Pharmacol. mechanical allodynia in rat models of neuropathic
2001;415:141–9. pain. Neurosci Res. 2009;63:42–6.
97. Jacus MO, Uebele VN, Renger JJ, Todorovic 113. Pichon X, Wattiez AS, Becamel C, et al. Disrupting
SM.  Presynaptic Cav3.2 channels regulate excit- 5-HT(2A) receptor/PDZ protein interactions reduces
atory neurotransmission in nociceptive dorsal horn hyperalgesia and enhances SSRI efficacy in neuro-
neurons. J Neurosci. 2012;32:9374–82. pathic pain. Mol Ther. 2010;18:1462–70.
98. Obrosova IG.  Diabetic painful and insensate neu- 114. Vithlani M, Terunuma M, Moss SJ.  The dynamic
ropathy: pathogenesis and potential treatments. modulation of GABA(A) receptor trafficking and
Neurotherapeutics. 2009;6:638–47. its role in regulating the plasticity of inhibitory syn-
99. Sandireddy R, Yerra VG, Areti A, Komirishetty P, apses. Physiol Rev. 2011;91:1009–22.
Kumar A.  Neuroinflammation and oxidative stress 115. Kaila K, Price TJ, Payne JA, Puskarjov M, Voipio
in diabetic neuropathy: futuristic strategies based on J. Cation-chloride cotransporters in neuronal devel-
these targets. Int J Endocrinol. 2014;2014:674987. opment, plasticity and disease. Nat Rev Neurosci.
100. Andrabi SA, Kim NS, Yu SW, et  al. Poly(ADP-­ 2014;15:637–54.
ribose) (PAR) polymer is a death signal. Proc Natl 116. Bright DP, Renzi M, Bartram J, et  al. Profound
Acad Sci U S A. 2006;103:18308–13. desensitization by ambient GABA limits activation
101. Hewlings SJ, Kalman DS. Curcumin: a review of its of δ-containing GABAA receptors during spillover.
effects on human health. Foods. 2017;6:92. J Neurosci. 2011;31:753–63.
102. Acar A, Akil E, Alp H, et  al. Oxidative dam- 117. Delgado-Lezama R, Loeza-Alcocer E, Andrés
age is ameliorated by curcumin treatment in brain C, Aguilar J, Guertin PA, Felix R.  Extrasynaptic
and sciatic nerve of diabetic rats. Int J Neurosci. GABA(A) receptors in the brainstem and spi-
2012;122:367–72. nal cord: structure and function. Curr Pharm Des.
103. Zhao WC, Zhang B, Liao MJ, et al. Curcumin ame- 2013;19:4485–97.
liorated diabetic neuropathy partially by inhibition 118. Perez-Sanchez J, Lorenzo LE, Lecker I, et  al.
of NADPH oxidase mediating oxidative stress in the α5GABA. J Neurosci Res. 2017;95:1307–18.
spinal cord. Neurosci Lett. 2014;560:81–5.
The Spinal Cord in Diabetic Neuropathy 379

119. Loeza-Alcocer E, Canto-Bustos M, Aguilar J, 135. Moore KA, Kohno T, Karchewski LA, Scholz J,
González-Ramírez R, Felix R, Delgado-Lezama Baba H, Woolf CJ.  Partial peripheral nerve injury
R. α(5)GABA(A) receptors mediate primary affer- promotes a selective loss of GABAergic inhibition
ent fiber tonic excitability in the turtle spinal cord. J in the superficial dorsal horn of the spinal cord. J
Neurophysiol. 2013;110:2175–84. Neurosci. 2002;22:6724–31.
120. Bettler B, Kaupmann K, Mosbacher J, Gassmann 136. Malmberg AB, O’Connor WT, Glennon JC, Cesena
M.  Molecular structure and physiological func- R, Calcutt NA.  Impaired formalin-evoked changes
tions of GABA(B) receptors. Physiol Rev. of spinal amino acid levels in diabetic rats. Brain
2004;84:835–67. Res. 2006;1115:48–53.
121. Terunuma M.  Diversity of structure and function 137. Jolivalt CG, Lee CA, Ramos KM, Calcutt
of GABA.  Proc Jpn Acad Ser B Phys Biol Sci. NA.  Allodynia and hyperalgesia in diabetic rats
2018;94:390–411. are mediated by GABA and depletion of spi-
122. Kaupmann K, Schuler V, Mosbacher J, et al. Human nal potassium-chloride co-transporters. Pain.
gamma-aminobutyric acid type B receptors are dif- 2008;140:48–57.
ferentially expressed and regulate inwardly rec- 138. Coull JA, Beggs S, Boudreau D, et  al. BDNF
tifying K+ channels. Proc Natl Acad Sci U S A. from microglia causes the shift in neuronal anion
1998;95:14991–6. gradient underlying neuropathic pain. Nature.
123. Bohlhalter S, Weinmann O, Mohler H, Fritschy 2005;438:1017–21.
JM.  Laminar compartmentalization of GABAA-­ 139. Lee-Kubli CA, Calcutt NA. Altered rate-dependent
receptor subtypes in the spinal cord: an immunohis- depression of the spinal H-reflex as an indicator of
tochemical study. J Neurosci. 1996;16:283–97. spinal disinhibition in models of neuropathic pain.
124. Roberts LA, Beyer C, Komisaruk BR.  Nociceptive Pain. 2014;155:250–60.
responses to altered GABAergic activity at the spinal 140. Fernyhough P, Diemel LT, Brewster WJ, Tomlinson
cord. Life Sci. 1986;39:1667–74. DR.  Altered neurotrophin mRNA levels in periph-
125. Yaksh TL.  Behavioral and autonomic correlates of eral nerve and skeletal muscle of experimentally dia-
the tactile evoked allodynia produced by spinal gly- betic rats. J Neurochem. 1995;64:1231–7.
cine inhibition: effects of modulatory receptor sys- 141. Lee-Kubli C, Marshall AG, Malik RA, Calcutt
tems and excitatory amino acid antagonists. Pain. NA. The H-reflex as a biomarker for spinal disinhi-
1989;37:111–23. bition in painful diabetic neuropathy. Curr Diab Rep.
126. Sivilotti L, Woolf CJ. The contribution of GABAA 2018;18:1.
and glycine receptors to central sensitization: dis- 142. Wager EW, Buerger AA.  A linear relationship
inhibition and touch-evoked allodynia in the spinal between H-reflex latency and sensory conduc-
cord. J Neurophysiol. 1994;72:169–79. tion velocity in diabetic neuropathy. Neurology.
127. Sorkin LS, Puig S, Jones DL.  Spinal bicuculline 1974;24:711–4.
produces hypersensitivity of dorsal horn neurons: 143. Stanley EF.  Sensory and motor nerve conduction
effects of excitatory amino acid antagonists. Pain. velocities and the latency of the H reflex during
1998;77:181–90. growth of the rat. Exp Neurol. 1981;71:497–506.
128. Wilson PR, Yaksh TL.  Baclofen is antinociceptive 144. Cliffer KD, Tonra JR, Carson SR, et al. Consistent
in the spinal intrathecal space of animals. Eur J repeated M- and H-Wave recording in the hind limb
Pharmacol. 1978;51:323–30. of rats. Muscle Nerve. 1998;21:1405–13.
129. Hammond DL, Drower EJ.  Effects of intrathecally 145. Palmieri RM, Ingersoll CD, Hoffman MA.  The
administered THIP, baclofen and muscimol on noci- hoffmann reflex: methodologic considerations and
ceptive threshold. Eur J Pharmacol. 1984;103:121–5. applications for use in sports medicine and athletic
130. Miletic G, Draganic P, Pankratz MT, Miletic training research. J Athl Train. 2004;39:268–77.
V.  Muscimol prevents long-lasting potentiation of 146. Lloyd DP, Wilson VJ. Reflex depression in rhythmi-
dorsal horn field potentials in rats with chronic con- cally active monosynaptic reflex pathways. J Gen
striction injury exhibiting decreased levels of the Physiol. 1957;40:409–26.
GABA transporter GAT-1. Pain. 2003;105:347–53. 147. Ishikawa K, Ott K, Porter RW, Stuart D.  Low fre-
131. Gwak YS, Tan HY, Nam TS, Paik KS, Hulsebosch quency depression of the H wave in normal and spi-
CE, Leem JW. Activation of spinal GABA receptors nal man. Exp Neurol. 1966;15:140–56.
attenuates chronic central neuropathic pain after spi- 148. Meinck HM.  Occurrence of the H reflex and
nal cord injury. J Neurotrauma. 2006;23:1111–24. the F wave in the rat. Electroencephalogr Clin
132. Basbaum AI, Bautista DM, Scherrer G, Julius Neurophysiol. 1976;41:530–3.
D. Cellular and molecular mechanisms of pain. Cell. 149. Schindler-Ivens S, Shields RK.  Low frequency
2009;139:267–84. depression of H-reflexes in humans with acute
133. Costigan M, Scholz J, Woolf CJ. Neuropathic pain: a and chronic spinal-cord injury. Exp Brain Res.
maladaptive response of the nervous system to dam- 2000;133:233–41.
age. Annu Rev Neurosci. 2009;32:1–32. 150. Ho SM, Waite PM.  Effects of different anesthetics
134. Castro-Lopes JM, Tavares I, Coimbra A.  GABA on the paired-pulse depression of the h reflex in adult
decreases in the spinal cord dorsal horn after periph- rat. Exp Neurol. 2002;177:494–502.
eral neurectomy. Brain Res. 1993;620:287–91.
380 A. G. Marshall et al.

151. Kakinohana O, Hefferan MP, Nakamura S, et  al. altered by long-term diabetes in epineurial arterioles
Development of GABA-sensitive spasticity and of the rat sciatic nerve. Diabetes Metab Res Rev.
rigidity in rats after transient spinal cord isch- 2006;22:211–9.
emia: a qualitative and quantitative electrophysi- 165. Yorek MA, Coppey LJ, Gellett JS, Davidson
ological and histopathological study. Neuroscience. EP.  Sensory nerve innervation of epineurial arteri-
2006;141:1569–83. oles of the sciatic nerve containing calcitonin gene-­
152. Lee-Kubli CA, Zhou X, Jolivalt CG, Calcutt related peptide: effect of streptozotocin-induced
NA. Pharmacological modulation of rate-dependent diabetes. Exp Diabesity Res. 2004;5:187–93.
depression of the spinal H-reflex predicts thera- 166. Schechtmann G, Song Z, Ultenius C, Meyerson BA,
peutic efficacy against painful diabetic neuropathy. Linderoth B.  Cholinergic mechanisms involved in
Diagnostics (Basel). 2021;11:283. the pain relieving effect of spinal cord stimulation
153. Hernandez-Reyes JE, Salinas-Abarca AB, Vidal-­ in a model of neuropathy. Pain. 2008;139:136–45.
Cantu GC, et  al. alpha5GABAA receptors play a 167. Stiller CO, Cui JG, O'Connor WT, Brodin E,
pronociceptive role and avoid the rate-dependent Meyerson BA, Linderoth B.  Release of gamma-­
depression of the Hoffmann reflex in diabetic neuro- aminobutyric acid in the dorsal horn and suppres-
pathic pain and reduce primary afferent excitability. sion of tactile allodynia by spinal cord stimulation in
Pain. 2019;160:1448–58. mononeuropathic rats. Neurosurgery. 1996;39:367–
154. Marshall AG, Lee-Kubli C, Azmi S, et al. Spinal dis- 74; discussion 374-365.
inhibition in experimental and clinical painful dia- 168. de Vos CC, Rajan V, Steenbergen W, van der Aa
betic neuropathy. Diabetes. 2017;66:1380–90. HE, Buschman HP. Effect and safety of spinal cord
155. Sultan A, Gaskell H, Derry S, Moore RA. Duloxetine stimulation for treatment of chronic pain caused
for painful diabetic neuropathy and fibromyalgia by diabetic neuropathy. J Diabetes Complications.
pain: systematic review of randomised trials. BMC 2009;23:40–5.
Neurol. 2008;8:29. 169. Pluijms WA, Slangen R, Joosten EA, et al. Electrical
156. Melzack R, Wall PD. Pain mechanisms: a new the- spinal cord stimulation in painful diabetic polyneu-
ory. Science. 1965;150:971–9. ropathy, a systematic review on treatment efficacy
157. Vannemreddy P, Slavin KV. Spinal cord stimulation: and safety. Eur J Pain. 2011;15:783–8.
current applications for treatment of chronic pain. 170. Slangen R, Schaper NC, Faber CG, et  al. Spinal
Anesth Essays Res. 2011;5:20–7. cord stimulation and pain relief in painful dia-
158. McGreevy K, Williams KA. Contemporary insights betic peripheral neuropathy: a prospective two-­
into painful diabetic neuropathy and treatment with center randomized controlled trial. Diabetes Care.
spinal cord stimulation. Curr Pain Headache Rep. 2014;37:3016–24.
2012;16:43–9. 171. van Beek M, Geurts JW, Slangen R, et al. Severity
159. McCarthy KF, McCrory C. Cerebrospinal fluid lev- of neuropathy is associated with long-term spi-
els of glial cell-derived neurotrophic factor correlate nal cord stimulation outcome in painful diabetic
with spinal cord stimulation frequency in patients peripheral neuropathy: five-year follow-up of a
with neuropathic pain: a preliminary report. Spinal prospective two-center clinical trial. Diabetes Care.
Cord. 2014;52(Suppl 2):S8–10. 2018;41:32–8.
160. Wu M, Linderoth B, Foreman RD. Putative mecha- 172. Pluijms WA, van Kleef M, Honig WM, Janssen SP,
nisms behind effects of spinal cord stimulation on Joosten EA.  The effect of spinal cord stimulation
vascular diseases: a review of experimental studies. frequency in experimental painful diabetic polyneu-
Auton Neurosci. 2008;138:9–23. ropathy. Eur J Pain. 2013;17:1338–46.
161. Wu M, Komori N, Qin C, Farber JP, Linderoth B, 173. van Beek M, van Kleef M, Linderoth B, van Kuijk
Foreman RD.  Sensory fibers containing vanilloid SM, Honig WM, Joosten EA.  Spinal cord stimu-
receptor-1 (VR-1) mediate spinal cord stimulation-­ lation in experimental chronic painful diabetic
induced vasodilation. Brain Res. 2006;1107:177–84. polyneuropathy: delayed effect of high-frequency
162. Wu M, Komori N, Qin C, Farber JP, Linderoth B, stimulation. Eur J Pain. 2017;21:795–803.
Foreman RD.  Extracellular signal-regulated kinase 174. de Vos CC, Bom MJ, Vanneste S, Lenders MW,
(ERK) and protein kinase B (AKT) pathways de Ridder D.  Burst spinal cord stimulation evalu-
involved in spinal cord stimulation (SCS)-induced ated in patients with failed back surgery syndrome
vasodilation. Brain Res. 2008;1207:73–83. and painful diabetic neuropathy. Neuromodulation.
163. Wu M, Thorkilsen MM, Qin C, Farber JP, Linderoth 2014;17:152–9.
B, Foreman RD.  Effects of spinal cord stimula- 175. Tjepkema-Cloostermans MC, de Vos CC, Wolters
tion on peripheral blood circulation in rats with R, Dijkstra-Scholten C, Lenders MW.  Effect of
streptozotocin-induced diabetes. Neuromodulation. burst stimulation evaluated in patients familiar
2007;10:216–23. with spinal cord stimulation. Neuromodulation.
164. Davidson EP, Coppey LJ, Yorek MA.  Activity and 2016;19:492–7.
expression of the vanilloid receptor 1 (TRPV1) is
Brain Changes in Diabetes and
Cognitive Dysfunction

Geert Jan Biessels

The first reports on cognitive dysfunction in rela- norm referenced tests are considered as abnormal.
tion to diabetes date back a century ago [1]. Yet, In clinical research and care, acquired cognitive
only over the past two decades, the spectrum of impairment is generally classified with the diag-
cognitive changes occurring in people with dia- nostic labels “Mild Cognitive Impairment (MCI)”
betes in terms of clinical features and prognosis or dementia. MCI refers to impairment affecting
has become more clear. While diabetes-­associated one or more cognitive domains with—by defini-
changes have been observed in all age groups, tion—largely preserved activities of daily life [4,
from young children to the oldest old, this chap- 5]. This construct was devised over two decades
ter will primarily discuss findings in adults. For ago to capture a stage between normal cognition
cognitive changes that are observed in children and dementia [5]. Individuals with MCI are at risk
with diabetes, the reader is referred to dedicated of transition to dementia, but not all people with
reviews [2, 3]. MCI show cognitive decline and in some cogni-
tion may even revert back to normal. Dementia is
defined as acquired objective cognitive impair-
1 Stages of Cognitive ment affecting multiple cognitive domains, severe
Dysfunction: Definitions enough to affect activities of daily life [6]. Most
people with dementia will show cognitive decline,
When considering different stages of cognitive although this clearly depends on the aetiology. It
dysfunction, it is essential to discern cognitive is important to note that neither the definition of
impairment from more subtle cognitive changes. MCI nor that of dementia refer to any particular
The term cognitive impairment is best reserved for underlying aetiology. Yet, for lay people, as well
acquired cognitive dysfunction where the actual as for many clinicians, a diagnosis of dementia is
cognitive performance on neuropsychological often implicitly equated with “Alzheimer’s dis-
testing falls outside the normal range. While there ease”. Although Alzheimer’s disease is indeed the
is no universally accepted definition of “abnormal most common form of dementia, it is important to
test results” among neuropsychologist, usually realize that MCI or dementia is principally syn-
scores below the 5th to the 10th percentile on dromal diagnosis, defined by symptoms. Such
symptoms may be suggestive of particular demen-
tia aetiologies, but are known to lack specificity.
G. J. Biessels (*) Therefore, the current standard in research—and
Department of Neurology G03.232, UMC Utrecht
increasingly also in clinical care—is to diagnose
Brain Center, University Medical Center Utrecht,
Utrecht, the Netherlands underlying aetiologies with dedicated biomarkers
e-mail: g.j.biessels@umcutrecht.nl for the actual disease process [7]. Such biomarker
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 381
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_21
382 G. J. Biessels

approaches have also shown that the pathologies definition be preserved [15]. Longitudinal studies
ultimately contributing to the development of show that diabetes-­ associated cognitive decre-
dementia may start to develop many years before ments show only slow progression over years [15,
cognitive symptoms develop. Alzheimer pathol- 16]. Because the construct “diabetes-associated
ogy, for example, may occur in individuals who cognitive decrements” refers to cognitive changes
are cognitively normal already two decades before that can be detected at a group level, it cannot be
dementia becomes clinically manifest [8, 9]. Yet, readily used as a diagnostic label in individual
as long as treatment for most types of dementia is patients. Yet, it may serve to understand cognitive
primarily symptomatic and supportive, it can be complaints expressed by an individual patient if
argued that for clinical practice, biomarker-sup- the severity and trajectories of the deficits fit with
ported diagnosis is not essential. Clearly, this is the construct’s definition.
different for aetiological research. For example, it In my view, diabetes-associated cognitive dec-
is important to be aware that approximately one in rements, on the one hand, and MCI and demen-
six people who have a clinical diagnosis of “prob- tia, on the other, should not be regarded as a
able Alzheimer’s disease”, based on a syndromal continuum [16]. These different stages of cogni-
diagnosis without biomarker support [10], do not tive dysfunction affect different age groups, have
have biomarker or pathological evidence of under- different trajectories, and appear to have different
lying Alzheimer pathology [11, 12]. This is clearly underlying mechanisms [17]. Diabetes-associated
relevant for studies of dementia occurring in asso- decrements can occur in all age groups, from
ciation with diabetes, as will be described below, young adults and even adolescents with T2DM
as most epidemiological studies on that topic [18, 19] to the oldest old [20]. Cognitive decline
based their diagnoses on dementia types on syn- over time is typically slow over the course of
dromal, rather than biomarker supported, many years [16]. These decrements affect people
diagnoses. with diabetes as a group: they are characterized
While diagnostic constructs for cognitive by a modest shift in average cognitive perfor-
impairment are by nature dichotomous classifiers, mance across individuals, rather than abnormal
which are present or absent in an individual performance in a subset of individuals. Dementia
patient, the more subtle cognitive changes that and MCI, on the other hand, are defined by abnor-
occur in association with diabetes rather present mal cognitive performance in individual patients.
themselves as a continuum. It has been noted that Occurrence of dementia and MCI is strongly age-­
as a group, people with diabetes perform on aver- dependent [21], and particularly dementia mostly
age slightly worse on cognitive testing than people involves relentless further cognitive decline.
without diabetes. This has been noted across all Clearly, most people who develop MCI or
age groups, for people with both type 1 and type 2 dementia will generally pass through a stage of
diabetes [13, 14]. The magnitude of the group dif- cognitive performance deficits that equate to cog-
ference is typically around 0.3–0.5 standard devia- nitive decrements. Yet, there are really no argu-
tion units [13, 14]. In other words, mean ments to generically consider all people with
performance on the domains that are most typi- diabetes-associated cognitive decrements to be
cally affected by diabetes is at the 40th to the 30th on a path towards dementia.
percentile of that of reference groups of people
without diabetes. This is well above the threshold
of the 5th to 10th percentile that denote cognitive 2 Stages of Cognitive
impairment. These subtle cognitive changes have Dysfunction in Diabetes
been coined “diabetes-associated cognitive decre-
ments” [15]. The subtle changes in cognitive func- 2.1 Type 1 Diabetes
tion to which this term refers might result in
cognitive complaints (usually expressed only by At a group level, adults with T1DM present with
the patient), but activities of daily life should by subtle cognitive decrements relative to age-­
Brain Changes in Diabetes and Cognitive Dysfunction 383

matched controls [13, 22]. These decrements par- literature on this topic. A retrospective cohort
ticularly affect the domain intelligence (effect study of administrative hospital records from
size Cohen’s d  =  0.7), psychomotor efficiency England observed that the relative risk for demen-
(d = 0.6), and cognitive flexibility (d = 0.5) [13] tia in people admitted with type 1 diabetes was
(see also Table  1). Long-term follow-up of the 1.65 (95% CI 1.61, 1.68), compared to a refer-
participants of the Diabetes Control and ence group [25]. This increased risk included
Complications Trial (DCCT) shows that cogni- both a syndromal diagnosis of Alzheimer’s dis-
tive decline over time is generally limited up to a ease (relative risk 1.40; 95% CI 1.27, 1.53) and
diabetes duration of ~25  years, although at an vascular dementia (2.66; 95% CI 2.48, 2.86).
average diabetes duration of 37 years, substantial Although the relative risk of dementia was high-
deterioration of psychomotor and mental effi- est in people with type 1 diabetes admitted to
ciency was observed [23]. There may also be hospital when aged 30–39  years (7.10; 95% CI
subgroups of patients, particularly those with 4.65, 10.6) and decreased as age increased, for
advanced microvascular complications, who may example to 1.61 (95% CI 1.57, 1.66) in those
show more pronounced cognitive decline over aged 70–79 years, the absolute risk of dementia
time [24]. Risk factors for accelerated cognitive occurrence was clearly much lower in the young-
decline in people with type 1 diabetes also est age band than in older people (30–39 years:
include exposure to higher HbA1c levels, more 1.0/1000; 50–59  years: 8.5/1000; 70–79  years:
episodes of severe hypoglycaemia, and elevated 59.2/1000) [25], showing that also among people
systolic blood pressure [23]. with diabetes occurrence of dementia below the
With increasing numbers of people with type age of 60  years is relatively rare. Exposure to
1 diabetes reaching old age, the question if type 1 higher HbA1c levels [26], episodes of severe
diabetes increases the risk of dementia becomes hypo- or hyperglycaemia [27], and presence of
more and more relevant, and there is an emerging diabetic retinopathy [28] or depression [29] have
been identified as risk factors for dementia among
people with type 1 diabetes, but this topic war-
Table 1  Cognitive decrements in type 1 and type 2
rants further investigation.
diabetes
Cognitive domain Type 1 Type 2
General cognition 0.2– 0.3
0.3 2.2 Type 2 Diabetes
Attention 0–0.3 0.2–
0.4 Subtle cognitive decrements in people with type
Psychomotor speed 0.3– 0.3 2 diabetes can be seen across cognitive domains,
0.6 with little specificity in terms of domains affected
Verbal memory 0–0.2 0.2–
(Table  1). Relative to people without diabetes,
0.3
Non-verbal memory 0–0.2 0.2– effect sizes are mostly small to moderate (Cohen’s
0.3 d 0.2–0.3) [14, 30]. Numerous studies have noted
Mental flexibility—executive 0.4– 0.3 these decrements in patients with type 2 diabetes
function 0.6 of all age groups [14, 30]. Onset is likely already
Visuospatial 0.4 0.2 in pre-diabetic stages [16]. Further diabetes-­
Estimates of the difference in average performance in related cognitive decline generally evolves only
people with diabetes compared to age-matched reference
groups per domain expressed in standardized effect sizes
slowly over time, at a rate that is up to 50% faster
(Cohen’s d; i.e. the difference between the means of two than that of normal cognitive ageing [16, 31, 32].
groups divided by the pooled standard deviation of the Population-based studies report an increased
two groups), based on meta-analyses by Brands [13], risk of MCI in people with type 2 diabetes, com-
Palta [14], and Monette [30] et al. In neuropsychological
studies, an effect size of 0.2 is generally considered to cor-
pared to people without diabetes (e.g. Ref. [33]).
respond to small effects, 0.5 to medium, and 0.8 to large A recent meta-analysis estimated the relative risk
effects [106] at 1.49 (95% CI 1.26–1.77) [34]. This included
384 G. J. Biessels

both so-called amnestic MCI (where the princi- tional attainment [37, 38]. Moreover, among
pally affected domain is memory; relative risk patients with type 2 diabetes episodes of severe
1.50; 95% CI 1.17–1.92) and non-amnestic MCI hypo- or hyperglycaemia, presence of micro- or
(where another non-memory domain is princi- macrovascular complications, including stroke,
pally affected; relative risk 1.34; 95% CI 1.04– and a low body mass index are known risk factors
1.73) [34]. The MCI risk appears to be higher in for dementia [39, 40]. The relation between gly-
those with higher HbA1c levels (i.e. HbA1c ≥7%, cemia and dementia risk in people with type 2
compared with HbA1c <7%, hazard ratio 1.56 diabetes is not completely clear [41]. There are
(95% CI 1.10–2.21) [33]. There is also consistent indications that both high and low HbA1c levels
evidence that type 2 diabetes increases the rate of may negatively impact cognition particularly in
conversion from MCI to dementia. The odds ratio older individuals [42], as has also been noted in
was estimated at 1.53 (95% CI 1.20–1.97) in a type 1 diabetes [26]. There are also indications
recent meta-analysis of ten studies [35], whereas that in people without diabetes fasting plasma
another meta-analysis of nine studies found a rel- glucose is related to dementia risk [34]. More
ative risk of 1.91 (95%CI 1.54–2.36) [34]. recently, however, evidence form Mendelian ran-
There have also been many studies on the risk domization studies questions if glucose levels
of dementia in relation to diabetes [34, 36]. and even type 2 diabetes itself (as defined by
Although not all of these studies formally distin- abnormal glucose levels) are causally related to
guished between type 1 and type 2 diabetes, the dementia, in particular Alzheimer’s disease [43,
vast majority of the patients involved will have 44]. In the light of these latter observations it
had type 2 diabetes [36]. A recent meta-analysis, been suggested that type 2 diabetes essentially
pooling 31 studies, with a total sample size of represents a risk marker for dementia and that the
4.3  million, showed a relative risk of all-cause excess dementia risk in people with type 2 diabe-
dementia of 1.43 (95% CI 1.33–1.53). Meta-­ tes primarily reflects lower socioeconomic posi-
regression analyses showed that the degree to tion and childhood and educational attainment
which cardiovascular disease was controlled for [45]. On the other hand, Mendelian randomiza-
explained heterogeneity between studies: the tion studies do suggest that type 2 diabetes is
pooled relative risk of studies not controlling for associated with small vessel disease-related brain
cardiovascular disease was 1.66 (95% CI 1.53– injury [46]. All in all these combined observa-
1.80), compared to 1.28 (95% CI 1.20–1.36) in tions suggest that the interrelations between type
studies controlling for cardiovascular disease 2 diabetes and dementia are complex, likely
[34]. Twenty-four studies included in the meta-­ depending on multiple factors, in part with non-­
analysis reported data for diabetes and the risk of linear relationships, and—as will be discussed
a clinical diagnosis of Alzheimer’s disease, with further on in this chapter—likely depending on
a relative risk of 1.43 (95% CI 1.25–1.62) [34]. multiple underlying disease processes.
Seventeen studies reported on the relative risk of
vascular dementia, which was estimated at 1.91
(95% CI 1.61–2.25) [34]. Importantly, diagnoses 3 Mechanisms: Structural
of Alzheimer’s disease in the studies included in Brain Changes and Dementia
the meta-analysis relied on a syndromal diagno- Aetiologies
sis, rather than biomarkers, which is known to
have limited specificity for the actual presence of Studying the aetiology and underlying brain
Alzheimer’s pathology [7, 11]. changes of cognitive dysfunction and dementia
Risk factors for dementia for dementia in the has long relied primarily on post-mortem neuro-
general population include several factors that pathological examination. Fortunately, over the
are closely related to type 2 diabetes, including past decades new neuroimaging methods and
midlife hypertension, midlife obesity, physical other biomarkers have been established that pro-
inactivity, depression, smoking, and low educa- vide much more options for mechanistic studies,
Brain Changes in Diabetes and Cognitive Dysfunction 385

importantly, also during life. This has proven to total tau and phosphorylated tau (P-tau) levels
be of great value for understanding the impact of can be measured in cerebrospinal fluid (CSF)
diabetes on the brain [47]. The plethora of differ- samples, obtained by lumbar puncture. These
ent markers can, however, provide challenges protein levels in the CSF provide an indication of
with interpretation. A key element in this context the burden of Aβ and tau pathologies in the brain
is to consider biomarker specificity for the pro- [7]. Evaluation of plasma levels of Aβ and tau
cess one intends to measure. For example, if one proteins as markers of Alzheimer pathologies is
is interested in detecting Alzheimer-related brain still in development [50].
changes, a conventional approach has long been Despite the extensive epidemiological evi-
to look at atrophy of the hippocampus, because dence for an association between diabetes and
atrophy in this region is closely linked to the risk of a syndromal diagnosis of Alzheimer’s
Alzheimer’s disease. The point is, however, that disease, post-mortem neuropathological studies
hippocampal atrophy is not specific to Alzheimer’s do not observe a higher burden of Aβ plaques and
disease [7]. Hence, if one truly wishes to assess tau tangles in people with diabetes compared to
Alzheimer’s pathology in relation to diabetes, it is those without diabetes [51, 52]. Moreover, PET
preferred to use currently available biomarkers for imaging studies show no influence of diabetes on
ß-amyloid (Aß) deposition or pathologic tau, as Aβ deposition [47, 53]. For example, in a pro-
these proteins are the constituents of the plaques spective population-based cohort study, neither
and tangles that define Alzheimer’s disease as a mid-life nor late-life diabetes was associated
unique disease among several that can lead to with abnormal Aβ tracer uptake on PET scans
dementia [7]. To support design of further bio- acquired in late-life [53]. This suggests that the
marker studies in diabetes and facilitate interpre- excess dementia risk in people with diabetes is
tation, we have recently proposed a framework to likely attributable to non-Alzheimer pathologies.
arrange them in three broad classes [47]. Firstly,
biomarkers reflecting pathologies considered to
define major causes of dementia, in particular 3.2 Diabetes and Vascular Brain
Alzheimer’s and vascular disease. Secondly, bio- Injury
markers of brain parenchymal injury that may
occur as a consequence of different disease pro- It is important to realize that most methods to
cesses. Thirdly, biomarkers of brain blood flow assess possible vascular contributions to cognitive
and metabolism, abnormalities of which may con- decline and dementia involve assessment of
tribute to brain injury. We made the point that parenchymal brain lesions that are the conse-
although biomarkers of brain parenchymal injury quence of cerebrovascular disease. This is because
may show patterns of injury suggestive of certain the primary vessel populations involved are so
aetiologies, caution is warranted to derive aetiol- small that they cannot be examined by conven-
ogy from such markers, because of limited aetio- tional neuroimaging techniques. Manifestations
logical specificity. Moreover, while abnormal of these so-called cerebral small vessel diseases
patterns of brain blood flow and metabolism may (SVDs) in the form of brain parenchymal lesions
reflect processes that can cause further brain can be detected both with CT and MRI, although
injury, brain injury by itself can affect cerebral sensitivity of MRI is superior [54]. The principal
blood flow and metabolism [47]. manifestations on MRI include lacunes, white
matter hyperintensities, microbleeds, microin-
farcts and enlarged perivascular spaces (Figs.  1
3.1 Diabetes and Alzheimer and 2) [54]. There are also emerging techniques to
Pathologies assess SVDs at the level of the small cerebral
blood vessels themselves [55], but at the time of
Brain Aβ and tau load can be imaged using PET writing these have not yet been evaluated in rela-
technology [48, 49]. In addition Aβ42, Aβ40, tion to diabetes.
386 G. J. Biessels

SVDs are common, and the main vascular 6.7% of controls [59]. As expected at this age, the
aetiology in dementia [56, 57] and vascular dis- burden of white matter hyperintensities was
ease is a key contributor to other diabetic compli- mostly low (Fazekas grade 1 [60], see Fig. 1) and
cations. Evaluation of SVD-related brain injury only 2.1% of patients and 0 controls had lacunes,
is therefore of clear interest for diabetes-­ too few to permit formal statistical testing [59].
associated cognitive dysfunction [58]. Another study, including 97 patients (average
For type 1 diabetes, few neuroimaging studies diabetes duration 41 years) and 81 controls, aver-
have thus far have reported on SVD lesions. This age age 50 years, did observe a higher burden of
is likely due to the fact that most brain MRI stud- white matter hyperintensities, with 33% of
ies of patients with type 1 diabetes included pop- patients having a Fazekas score of 2 or 3, com-
ulations under the age of 40–50 years, and sample pared to 7% of controls, with 5% of patients
sizes were generally modest. Because SVD showing lacunes versus none of the controls [61].
lesions typically are relatively rare or low in bur- Among patients with type 1 diabetes, micro-
den at that age, this limits the power of such stud- bleeds were found to be more common in those
ies. A study with the largest sample size thus far with than in those without proliferative retinopa-
(191 patients, 30 matched controls, average age thy [62].
40  years) did observe that microbleeds were The literature on vascular brain lesions in
seven times as common in patients (24%) as con- patients with type 2 diabetes is more extensive
trols (3.3%), and some form of white matter [63, 64]. Autopsy studies report a more frequent
hyperintensities was seen in 23% of patients and occurrence of brain infarcts in people with type

Fig. 1  Grading white matter hyperintensities. White mat- deep white matter. Deep (subcortical) WMH are graded
ter hyperintensities (WMH) on Fluid Attenuated Inversion separately as: 0 = absence, 1 = punctuate foci, 2 = begin-
Recovery (FLAIR) or other T2-weighted sequences can ning confluence of foci, and 3  =  large confluent areas.
be rated visually according to the Fazekas scale [60]. Examples of each grade are provided. The patient with
Periventricular WMHs are graded as: 0  =  absence, grade 3 WMH also has multiple lacunes, examples marked
1  =  caps or pencil-thin lining, 2  =  smooth halo, and by white arrowheads
3  =  irregular periventricular WMH extending into the
Brain Changes in Diabetes and Cognitive Dysfunction 387

a b

c d

Fig. 2  Examples of vascular brain injury on MRI. (a) deep, strictly lobar, and mixed (as is the case in this
Right frontal lacune (white arrowhead). On FLAIR patient). It is important to distinguish microbleeds from
images lacunes of presumed vascular origin generally other sources of signal void such as blood vessels (open
have a central CSF-like cystic core with a surrounding rim arrowheads) and mineral deposits in the basal ganglia
of hyperintensity [54]. This patient also has Fazekas grade (star), based on shape and location. (c/d) An acute cortical
3 WMH [60]. (b) Cerebral microbleeds (white arrow- infarct showing up as a high signal on FLAIR (c) with a
heads) on a T2*-weighted image. Microbleeds appear as corresponding lesion on diffusion MRI (d). This patient
areas of signal void on T2*-weighted MRI sequences also has Fazekas grade 3 WMH (c)
[54]. They can be rated according to location as strictly
388 G. J. Biessels

2 diabetes compared to people without diabetes Studies on brain volumes in people with type
[64]. These studies also report that vessel 1 diabetes generally have modest sample sizes. A
pathology is more common in those with type 2 meta-analysis of volumetric data found no sig-
diabetes, in particular cerebral atherosclerosis nificant differences in total brain volume, total
[64]. By contrast, type 2 diabetes does not grey matter volume, or hippocampal volume
appear to be associated with cerebral arteriolo- between people with type 1 and controls [73].
sclerosis at autopsy [64, 65]. This requires fur- Some studies do show regional differences in
ther investigation and seems counterintuitive, in cortical grey matter volume or density within the
the light of type 2 diabetes’ known association frontal [74–76], posterior [76, 77], and temporal
with peripheral arteriolosclerosis and with sub- cortex [76]. The patterns of affected regions vary
cortical infarcts in the brain [64]. Indeed, type 2 across studies, likely reflecting the subtle nature
diabetes is an established risk factor for lacunes of the changes, variation in study selection crite-
on CT and MRI [63, 66, 67], and Mendelian ria, and modest sample sizes. There are indica-
randomization analyses also suggest a causal tions that lower volumes are associated with
association between type 2 diabetes and lacunar worse glycaemic control [76, 77] and also with
stroke [68]. The volume of white matter hyper- worse cognitive performance [77], but these top-
intensities also appears to be modestly increased ics warrant further studies with larger sample
in people with type 2 diabetes, compared to sizes, also including people with more longstand-
those without [17, 67, 69]. White matter hyper- ing diabetes.
intensities appear to be higher in patients with Modest brain atrophy, cortical as well as sub-
higher HbA1c levels [70]. There is as yet no cortical, is a consistent finding of MRI studies
convincing evidence that type 2 diabetes affects in type 2 diabetes. Cross-sectional studies report
the burden of microbleeds, microinfarcts and reductions in average total brain volume of 0.5–
enlarged perivascular spaces [17, 67]. While 2.0% relative to controls, which is the equiva-
occurrence of these different SVD lesions lent of 2–5 years of normal ageing [70, 73, 78].
appeared to be a mediator in type 2 diabetes- A meta-analysis reported regional reductions in
associated cognitive decrements on several cog- orbitofrontal cortex grey matter, the hippocam-
nitive domains [71], others found that the pus, and basal ganglia [73]. It is questionable,
association between type 2 diabetes and cogni- however, if hippocampal atrophy is really out of
tive impairment was not attenuated when SVD proportion to global atrophy [79]. Several [78,
lesions were taken into account [66]. 80], but not all [81], longitudinal studies show
that atrophy in people with type 2 diabetes
exceeds the rate of brain volume loss of normal
3.3 Diabetes ageing, although effects are modest. In this
and Neurodegeneration/ respect, the trajectories of brain volume changes
Global Brain Injury and those of diabetes-associated cognitive dec-
rements are much the same. Lower brain vol-
Modest reductions in cerebral volume, generally ume is the neuroimaging marker that most
considered to reflect cerebral atrophy, are the clearly differentiates patients with type 2 diabe-
most extensively documented neuroimaging fea- tes and MCI from those with type 2 diabetes
ture of type 1 as well as type 2 diabetes. Of note, without MCI [82].
atrophy can reflect underlying neurodegenerative Diffusion tensor imaging (DTI) is a marker of
disease, but it is also a manifestation of SVD brain parenchymal injury, primarily used to eval-
[54]. Moreover, for studies in diabetes, it should uate white matter changes [83]. The technique
be considered that fluctuations in blood glucose allows quantification of global white matter
levels can induce osmotic changes, which can injury, but can also be used to evaluate the integ-
also affect brain volumes [72], thus confounding rity of specific tract abnormalities and brain net-
atrophy assessment. work integrity [54, 63]. DTI studies in people
Brain Changes in Diabetes and Cognitive Dysfunction 389

with type 1 and type 2 diabetes show changes in autofluorescence [95]. In a large population-­
white matter microstructure and connectivity based cohort study, both higher levels of the cir-
compared to controls, which relate to cognitive culating AGEs skin autofluorescence were
dysfunction [67, 84, 85]. associated with worse cognitive performance in
Besides brain imaging, alternative biomarker people with and without type 2 diabetes [95].
approaches to detect neurodegeneration are Another study observed that higher skin autoflu-
emerging that may also be relevant for diabetes. orescence was associated with grey matter atro-
These include assessment of retinal nerve fibre phy in people with type 2 diabetes [96].
layer thickness with optical coherence tomogra-
phy [86] and blood-based markers such as neuro-
filament light [87, 88]. 4 Implications for Clinical
Practice

3.4 Markers of Cerebral Blood 4.1 Emerging Clinical Guidelines


Flow and Metabolism
Over the past years, several professional societies
Given the fact that diabetes is defined by meta- have published guidelines on the management of
bolic disturbances and is known to affect vascular cognitive impairment in relation to diabetes
function throughout the body, biomarkers of (Reviewed in Biessels and Whitmer [97]; see also
brain blood flow and metabolism are clearly of Text Box 1 [98–103]). Essentially, all of these
interest to explore the effects of diabetes on the guidelines recommend that cognitive impairment
brain. However, as mentioned earlier, a caveat in in people with diabetes:
the interpretation of abnormal patterns of cere-
bral blood flow and metabolism is that they may 1. Should be actively sought for (by screening)
represent both cause and effect of disease pro- 2. Dictates an individualized diabetes manage-
cesses in the brain. ment regimen, considering the patient’s capa-
Early studies, mostly with small sample sizes, bilities, generally with more lenient treatment
showed variable results on cerebral perfusion in targets and simplified treatment schemes to
people with diabetes [63, 89, 90]. More recent improve compliance and reduce treatment-­
studies, primarily involving using arterial spin related risks
labelling MRI, show that at least in people with
type 2 diabetes, global cerebral blood flow The rationale for these recommendations is
appears to be largely preserved [91]. Yet, there that among people with diabetes those with
are reports of regional flow reductions relative to ­cognitive impairment represent a risk group for
controls that also relate to worse cognitive per- poor health outcomes, that cognitive impairment
formance [92, 93]. hampers diabetes self-management, and that it is
Cerebral glucose metabolism is clearly also of related with a more frequent occurrence of treat-
interest in people with diabetes [94]. It is impor- ment related complications, in particular severe
tant, however, that inter-subject variation in blood hypoglycaemic episodes [97]. By screening for
glucose levels may affect tracer uptake in FDG-­ cognitive impairment and offering individualized
PET and may thus confound results [47]. care to those affected, these risks might be
Advanced glycation end-products (AGEs) mitigated.
result from nonenzymatic chemical reactions The emergence of this guidance is clearly
between reduced sugars and proteins. Formation important. Yet, implementation in clinical prac-
of AGEs is accelerated by hyperglycaemia [95]. tice, particularly with regard to routine screening
Circulating AGEs can be measured in plasma, but for cognitive impairment, does currently not
their accumulation in tissues can also be esti- appear to be widespread. Further development of
mated through measurement of so-called skin effective and efficient screening strategies that
390 G. J. Biessels

can be used in a diabetes care setting still required,


as well as further evidence that screening and Glycaemic Targets for Elderly
personalized management of cognitive impair- Patients with Diabetes Japan
ment in people with diabetes indeed improve Diabetes Society (JDS)/Japan
patient outcomes [97]. Geriatrics Society (JGS) 2016 [100]
• Individualized targets, with consideration
of age, severity of cognitive impairment
Text Box 1 Key Points from Clinical • Relatively higher glycaemic targets for
Guidelines on Screening, Diagnosis, and use of drugs potentially associated with
Clinical Management of Cognitive severe hypoglycaemia
Impairment Diabetes (Adapted from [97])
International Diabetes Federation American Diabetes Association
(IDF): Managing Older People with (ADA): “Standards of Medical Care
Type 2 Diabetes Global Guideline in Diabetes” 20191 [99]
2013 [98] • Annual screening for early detection of
• Assessment of older people with diabe- mild cognitive impairment or dementia
tes should be multidisciplinary, aiming is indicated for adults 65  years of age
to collect information on medical, psy- and older
chosocial, and functional capabilities • Older adults who are otherwise healthy
and how these may limit activities. with few coexisting chronic illnesses
Annual assessment with cognitive and intact cognitive function and func-
screening tests can be considered. tional status should have lower glycae-
Enquiry about functional capacity and mic goals (A1C<7.5% [58 mmol/mol]),
cognitive and mental health should be a while those with multiple coexisting
standard part of patient assessment. chronic illnesses, cognitive impairment,
• In patients with cognitive impairment or functional dependence should have
treatment targets should be more lenient, less stringent glycaemic goals
particularly for glycaemic control, (A1C < 8.0–8.5% [64–69 mmol/mol]).

Diabetes and Dementia in Older Endocrine Society: Treatment of


People: a Best Clinical Practice Diabetes in Older Adults 2019 [102]
Statement by a Multidisciplinary • Perform cognitive screening in patients
National (United Kingdom) Expert aged 65  years and older. Repeat every
Working Group 2014 [101] 2–3 years after a normal screening test
• Routinely employ a brief cognitive result for patients without cognitive
screening test in annual patient complaints or repeat 1 year after a bor-
assessments derline normal test result.
• In patients with cognitive impairment • In patients with a diagnosis of cognitive
therapy risk should be minimized: impairment (i.e. MCI or dementia), it is
suggested to simplify medication regi-
–– Avoid overly intensive management mens and tailor glycaemic targets (i.e.
–– Use therapies that reduce risk of less stringent).
hypoglycaemia
–– Focus education and support on both For full details see original publications.
patients and carers

The ADA guideline is an update from earlier recommen-


1 

dations [112].
Brain Changes in Diabetes and Cognitive Dysfunction 391

4.2 Diagnosing Cognitive complaints after 6–12  months is generally


Dysfunction in People recommended.
with Diabetes Patients suspected of the presence of MCI and
dementia clearly warrant another diagnostic
The optimal diagnostic strategy in people with approach [15, 104]. As there are no
diabetes and cognitive complaints depends on the ­diabetes-­specific features to MCI and dementia,
prior likelihood of presence of cognitive impair- the same diagnostic tests are indicated as in
ment. This likelihood is clearly strongly related patients without diabetes.
to age, but also determined by history taking.
Examples of “what went wrong” due to the cog-
nitive complaints can be helpful to estimate the 4.3 Treatment
likely severity of cognitive dysfunction. One
should collect information on impact on profes- As indicated above and summarized in Text Box
sional and social activities and on other aspects 1, current guidelines recommend tailored diabe-
of daily life. Trajectories of the complaints in tes treatment of people with diabetes and cogni-
terms of their onset and progression over time tive impairment.
should be determined. At present there are no treatments that can halt
In people below the age of 60  years, with cognitive decline due to neurodegenerative dis-
long-standing (i.e. several years) cognitive com- eases. Clearly, adequate cardiovascular risk fac-
plaints, that showed only slow progression over tor management is indicated, but this should be
time, with preserved social and professional guided by the generic diabetes management
activities, although possibly at the cost of a per- guidelines. In patients with manifestations of
ceived higher effort, presence of diabetes-­ vascular injury on MRI, but no history of stroke
associated cognitive decrements may be or other cardiovascular disease, primary preven-
considered. Accompanying fatigue is not uncom- tion strategies for prevention of cardiovascular
mon. Affective complaints should be evaluated disease generally apply. White matter hyperin-
and presence of depression should also be con- tensities as such, for example, are no solid indica-
sidered. In case of such subtle cognitive com- tion for prescription of antiplatelet drugs. The
plaints, ancillary investigations are mostly reader is referred to published recommendations
indicated to rule out alternative explanations. for the management of such “covert” cerebrovas-
Neuropsychological examination can be per- cular disease [105].
formed to formally quantify the severity of the At present there is no evidence that maintain-
cognitive deficits. Brief cognitive screening tests ing strict glycaemic control can prevent cognitive
are generally less useful in this setting, as they decline in people with diabetes [41, 106]. Avoiding
are not sensitive to subtle cognitive changes. severe hypoglycaemic episodes is important, but
Neuroimaging should be used on indication. that is already a cornerstone of regular diabetes
Routine MRI scanning of people with subtle cog- care. There is an expanding literature of class
nitive changes will likely yield a lot of chance effects of different glucose-­ lowering drugs on
findings, like low-grade white matter hyperinten- cognitive impairment, reporting benefit or poten-
sities or an occasional lacune. This may generate tial harm for different drugs [107, 108].
worries rather than comfort patients. Most impor- Importantly, most of the available data is from
tantly, it should be acknowledged that diabetes-­ observational studies and possibly subject by
associated cognitive decrements are not a formal bias, such as confounding by indication.
diagnostic label, for lack of specific criteria that Interestingly though, more and more head to head
can reliably pinpoint affected individuals. This comparisons of different drugs become available,
label should therefore be offered as a possible largely as an spin-off of the large cardiovascular
explanation for a patient’s complaints rather than outcome trials that have been conducted for these
a formal diagnosis [15]. Re-evaluation of the drugs over the past decades. While many of these
392 G. J. Biessels

trials have not reported benefit of one drug over 12. Kapasi A, DeCarli C, Schneider JA. Impact of mul-
tiple pathologies on the threshold for clinically overt
another (e.g. Refs [109, 110]), a recent study (the dementia. Acta Neuropathol. 2017;134(2):171–86.
REWIND trial) that compared the glucagon-like 13. Brands AM, Biessels GJ, de Haan EH, Kappelle
peptide-1 receptor agonist dulaglutide to placebo LJ, Kessels RP.  The effects of type 1 diabetes on
found an indication that dulaglutide was superior cognitive performance: a meta-analysis. Diabetes
­
Care. 2005;28(3):726–35.
in preventing cognitive impairment [111], 14. Palta P, Schneider AL, Biessels GJ, Touradji P, Hill-­
although it should be acknowledged that the two Briggs F.  Magnitude of cognitive dysfunction in
treatment arms in the trial also differed in terms of adults with type 2 diabetes: a meta-analysis of six
HbA1c levels. cognitive domains and the most frequently reported
neuropsychological tests within domains. J Int
Neuropsychol Soc. 2014;20(3):278–91.
15. Koekkoek PS, Kappelle LJ, van den Berg E, Rutten
References GE, Biessels GJ. Cognitive function in patients with
diabetes mellitus: guidance for daily care. Lancet
1. Miles WR, Root HF.  Psychologic tests applied in Neurol. 2015;14(3):329–40.
diabetic patients. Arch Intern Med. 1922;30:767–77. 16. Biessels GJ, Strachan MW, Visseren FL, Kappelle
2. Cameron FJ, Northam EA, Ryan CM.  The effect LJ, Whitmer RA.  Dementia and cognitive decline
of type 1 diabetes on the developing brain. Lancet in type 2 diabetes and prediabetic stages: towards
Child Adolesc Health. 2019;3(6):427–36. targeted interventions. Lancet Diabetes Endocrinol.
3. Cato A, Hershey T.  Cognition and type 1 diabe- 2014;2(3):246–55.
tes in children and adolescents. Diabetes Spectr. 17. Biessels GJ, Despa F. Cognitive decline and dementia
2016;29(4):197–202. in diabetes mellitus: mechanisms and clinical impli-
4. Petersen RC, Lopez O, Armstrong MJ, et al. Practice cations. Nat Rev Endocrinol. 2018;14(10):591–604.
guideline update summary: mild cognitive impair- 18. Brady CC, Vannest JJ, Dolan LM, et al. Obese ado-
ment: report of the guideline development, dis- lescents with type 2 diabetes perform worse than
semination, and implementation subcommittee of controls on cognitive and behavioral assessments.
the American Academy of Neurology. Neurology. Pediatr Diabetes. 2017;18(4):297–303.
2018;90(3):126–35. 19. Yates KF, Sweat V, Yau PL, Turchiano MM, Convit
5. Petersen RC. Mild cognitive impairment as a diag- A. Impact of metabolic syndrome on cognition and
nostic entity. J Intern Med. 2004;256(3):183–94. brain: a selected review of the literature. Arterioscler
6. American Psychiatric Association. Diagnostic and Thromb Vasc Biol. 2012;32(9):2060–7.
statistical manual of mental disorders, fourth edition 20. van den Berg E, de Craen AJ, Biessels GJ, Gussekloo
(DSM-IV). Washington DC: American Psychiatric J, Westendorp RG.  The impact of diabetes melli-
Association; 1994. tus on cognitive decline in the oldest of the old: a
7. Jack CR Jr, Bennett DA, Blennow K, et al. NIA-AA prospective population-based study. Diabetologia.
research framework: toward a biological defini- 2006;49(9):2015–23.
tion of Alzheimer’s disease. Alzheimers Dement. 21. Prince M, Bryce R, Albanese E, Wimo A, Ribeiro
2018;14(4):535–62. W, Ferri CP.  The global prevalence of dementia: a
8. Jansen WJ, Ossenkoppele R, Knol DL, et  al. systematic review and metaanalysis. Alzheimers
Prevalence of cerebral amyloid pathology in per- Dement. 2013;9(1):63–75 e2.
sons without dementia: a meta-analysis. JAMA. 22. Li W, Huang E, Gao S.  Type 1 diabetes mellitus
2015;313(19):1924–38. and cognitive impairments: a systematic review. J
9. Bateman RJ, Xiong C, Benzinger TL, et  al. Alzheimers Dis. 2017;57(1):29–36.
Clinical and biomarker changes in dominantly 23. Jacobson AM, Ryan CM, Braffett BH, et  al.
inherited Alzheimer’s disease. N Engl J Med. Cognitive performance declines in older adults with
2012;367(9):795–804. type 1 diabetes: results from 32 years of follow-
10. McKhann G, Drachman D, Folstein M, Katzman ­up in the DCCT and EDIC Study. Lancet Diabetes
R, Price D, Stadlan EM.  Clinical diagnosis of Endocrinol. 2021;9(7):436–45.
Alzheimer’s disease: report of the NINCDS-­ 24. Nunley KA, Rosano C, Ryan CM, et al. Clinically
ADRDA Work Group under the auspices of relevant cognitive impairment in middle-aged adults
Department of Health and Human Services with childhood-onset type 1 diabetes. Diabetes Care.
Task Force on Alzheimer’s Disease. Neurology. 2015;38(9):1768–76.
1984;34(7):939–44. 25. Smolina K, Wotton CJ, Goldacre MJ. Risk of demen-
11. Ossenkoppele R, Jansen WJ, Rabinovici GD, tia in patients hospitalised with type 1 and type 2
et  al. Prevalence of amyloid PET positivity in diabetes in England, 1998-2011: a retrospective
dementia syndromes: a meta-analysis. JAMA. national record linkage cohort study. Diabetologia.
2015;313(19):1939–49. 2015;58(5):942–50.
Brain Changes in Diabetes and Cognitive Dysfunction 393

26. Lacy ME, Gilsanz P, Karter AJ, Quesenberry CP, 40. Yu JH, Han K, Park S, et  al. Incidence and risk
Pletcher MJ, Whitmer RA. Long-term glycemic con- factors for dementia in type 2 diabetes mellitus:
trol and dementia risk in type 1 diabetes. Diabetes a nationwide population-based study in Korea.
Care. 2018;41(11):2339–45. Diabetes Metab J. 2020;44(1):113–24.
27. Whitmer RA, Gilsanz P, Quesenberry CP, Karter 41. Geijselaers SLC, Sep SJS, Stehouwer CDA, Biessels
AJ, Lacy ME.  Association of type 1 diabetes and GJ. Glucose regulation, cognition, and brain MRI in
hypoglycemic and hyperglycemic events and risk of type 2 diabetes: a systematic review. Lancet Diabetes
dementia. Neurology. 2021;97(3):e275–83. Endocrinol. 2015;3(1):75–89.
28. Rodill LG, Exalto LG, Gilsanz P, Biessels GJ, 42. Janssen J, van den Berg E, Zinman B, et al. HbA1c,
Quesenberry CP Jr, Whitmer RA. Diabetic retinopa- insulin resistance, and beta-cell function in rela-
thy and dementia in type 1 diabetes. Alzheimer Dis tion to cognitive function in type 2 diabetes: the
Assoc Disord. 2018;32(2):125–30. CAROLINA cognition substudy. Diabetes Care.
29. Gilsanz P, Schnaider Beeri M, Karter AJ, Quesenberry 2019;42(1):e1–3.
CP Jr, Adams AS, Whitmer RA. Depression in type 43. Larsson SC, Traylor M, Malik R, et al. Modifiable
1 diabetes and risk of dementia. Aging Ment Health. pathways in Alzheimer’s disease: Mendelian ran-
2019;23(7):880–6. domisation analysis. BMJ. 2017;359:j5375.
30. Monette MC, Baird A, Jackson DL. A meta-­analysis 44. Benn M, Nordestgaard BG, Tybjaerg-Hansen
of cognitive functioning in nondemented adults A, Frikke-Schmidt R.  Impact of glucose on
with type 2 diabetes mellitus. Can J Diabetes. risk of dementia: Mendelian randomisation
2014;38(6):401–8. studies in 115,875 individuals. Diabetologia.
31. Pappas C, Andel R, Infurna FJ, Seetharaman 2020;63(6):1151–61.
S.  Glycated haemoglobin (HbA1c), diabetes 45. James SN, Wong A, Tillin T, Hardy R, Chaturvedi
and trajectories of change in episodic memory N, Richards M. The effect of mid-life insulin resis-
performance. J Epidemiol Community Health. tance and type 2 diabetes on older-age cognitive
2017;71(2):115–20. state: the explanatory role of early-life advantage.
32. Yaffe K, Falvey C, Hamilton N, et  al. Diabetes, Diabetologia. 2019;62(10):1891–900.
glucose control, and 9-year cognitive decline 46. Liu J, Rutten-Jacobs L, Liu M, Markus HS, Traylor
among older adults without dementia. Arch Neurol. M.  Causal impact of type 2 diabetes mellitus on
2012;69(9):1170–5. cerebral small vessel disease: a Mendelian random-
33. Rawlings AM, Sharrett AR, Albert MS, et  al. The ization analysis. Stroke. 2018;49(6):1325–31.
association of late-life diabetes status and hyper- 47. Biessels GJ, Nobili F, Teunissen CE, Simo R,
glycemia with incident mild cognitive impairment Scheltens P.  Understanding multifactorial brain
and dementia: the ARIC study. Diabetes Care. changes in type 2 diabetes: a biomarker perspective.
2019;42(7):1248–54. Lancet Neurol. 2020;19(8):699–710.
34. Xue M, Xu W, Ou YN, et al. Diabetes mellitus and 48. Smith R, Scholl M, Leuzy A, et  al. Head-to-head
risks of cognitive impairment and dementia: a sys- comparison of tau positron emission tomography
tematic review and meta-analysis of 144 prospective tracers [(18)F]flortaucipir and [(18)F]RO948. Eur J
studies. Ageing Res Rev. 2019;55:100944. Nucl Med Mol Imaging. 2020;47(2):342–54.
35. Pal K, Mukadam N, Petersen I, Cooper C.  Mild 49. Herholz K, Ebmeier K.  Clinical amyloid imag-
cognitive impairment and progression to demen- ing in Alzheimer’s disease. Lancet Neurol.
tia in people with diabetes, prediabetes and 2011;10(7):667–70.
metabolic syndrome: a systematic review and 50. Palmqvist S, Janelidze S, Stomrud E, et  al.
meta-analysis. Soc Psychiatry Psychiatr Epidemiol. Performance of fully automated plasma assays
2018;53(11):1149–60. as screening tests for Alzheimer disease-related
36. Biessels GJ, Staekenborg S, Brunner E, Brayne C, beta-­amyloid status. JAMA Neurol. 2019;76(9):
Scheltens P. Risk of dementia in diabetes mellitus: a 1060–9.
systematic review. Lancet Neurol. 2006;5(1):64–74. 51. Abner EL, Nelson PT, Kryscio RJ, et al. Diabetes is
37. Norton S, Matthews FE, Barnes DE, Yaffe K, Brayne associated with cerebrovascular but not Alzheimer’s
C.  Potential for primary prevention of Alzheimer’s disease neuropathology. Alzheimers Dement.
disease: an analysis of population-based data. Lancet 2016;12(8):882–9.
Neurol. 2014;13(8):788–94. 52. Dos Santos Matioli MNP, Suemoto CK, Rodriguez
38. Deckers K, van Boxtel MP, Schiepers OJ, et  al. RD, et  al. Diabetes is not associated with
Target risk factors for dementia prevention: a sys- Alzheimer’s disease neuropathology. J Alzheimers
tematic review and Delphi consensus study on the Dis. 2017;60(3):1035–43.
evidence from observational studies. Int J Geriatr 53. Gottesman RF, Schneider AL, Zhou Y, et  al.
Psychiatry. 2015;30(3):234–46. Association between midlife vascular risk factors
39. Exalto LG, Quesenberry CP, Barnes D, Kivipelto and estimated brain amyloid deposition. JAMA.
M, Biessels GJ, Whitmer RA.  Midlife risk score 2017;317(14):1443–50.
for the prediction of dementia four decades later. 54. Wardlaw JM, Smith EE, Biessels GJ, et  al.
Alzheimers Dement. 2014;10(5):562–70. Neuroimaging standards for research into small ves-
394 G. J. Biessels

sel disease and its contribution to ageing and neu- brovascular disease on MRI: the Atherosclerosis Risk
rodegeneration. Lancet Neurol. 2013;12(8):822–38. in Communities Neurocognitive Study (ARIC-­NCS).
55. Zwanenburg JJM, van Osch MJP.  Targeting cere- Diabetes Care. 2017;40(11):1514–21.
bral small vessel disease with MRI.  Stroke. 71. Qiu C, Sigurdsson S, Zhang Q, et al. Diabetes, mark-
2017;48(11):3175–82. ers of brain pathology and cognitive function: the
56. O’Brien JT, Thomas A. Vascular dementia. Lancet. Age, Gene/Environment Susceptibility-Reykjavik
2015;386(10004):1698–706. Study. Ann Neurol. 2014;75(1):138–46.
57. Wardlaw JM, Smith C, Dichgans M.  Small ves- 72. Puri BK, Lewis HJ, Saeed N, Davey NJ. Volumetric
sel disease: mechanisms and clinical implications. change of the lateral ventricles in the human
Lancet Neurol. 2019;18(7):684–96. brain following glucose loading. Exp Physiol.
58. van Sloten TT, Sedaghat S, Carnethon MR, Launer 1999;84(1):223–6.
LJ, Stehouwer CDA.  Cerebral microvascular 73. Moulton CD, Costafreda SG, Horton P, Ismail K, Fu
complications of type 2 diabetes: stroke, cogni- CH.  Meta-analyses of structural regional cerebral
tive dysfunction, and depression. Lancet Diabetes effects in type 1 and type 2 diabetes. Brain Imaging
Endocrinol. 2020;8(4):325–36. Behav. 2015;9(4):651–62.
59. Thorn LM, Shams S, Gordin D, et al. Clinical and 74. Frokjaer JB, Brock C, Softeland E, et  al.
MRI features of cerebral small-vessel disease in type Macrostructural brain changes in patients with long-
1 diabetes. Diabetes Care. 2019;42(2):327–30. standing type 1 diabetes mellitus—a cortical thick-
60. Fazekas F, Chawluk JB, Alavi A, Hurtig HI, ness analysis study. Exp Clin Endocrinol Diabetes.
Zimmerman RA. MR signal abnormalities at 1.5 T 2013;121(6):354–60.
in Alzheimer’s dementia and normal aging. AJR Am 75. Hughes TM, Ryan CM, Aizenstein HJ, et al. Frontal
J Roentgenol. 1987;149(2):351–6. gray matter atrophy in middle aged adults with
61. Nunley KA, Ryan CM, Orchard TJ, et  al. White type 1 diabetes is independent of cardiovascular
matter hyperintensities in middle-aged adults risk factors and diabetes complications. J Diabetes
with childhood-onset type 1 diabetes. Neurology. Complications. 2013;27(6):558–64.
2015;84(20):2062–9. 76. Musen G, Lyoo IK, Sparks CR, et  al. Effects of
62. Woerdeman J, van Duinkerken E, Wattjes MP, type 1 diabetes on gray matter density as mea-
et al. Proliferative retinopathy in type 1 diabetes is sured by voxel-based morphometry. Diabetes.
associated with cerebral microbleeds, which is part 2006;55(2):326–33.
of generalized microangiopathy. Diabetes Care. 77. Franc DT, Kodl CT, Mueller BA, Muetzel RL,
2014;37(4):1165–8. Lim KO, Seaquist ER.  High connectivity between
63. Brundel M, Kappelle LJ, Biessels GJ.  Brain imag- reduced cortical thickness and disrupted white mat-
ing in type 2 diabetes. Eur Neuropsychopharmacol. ter tracts in long-standing type 1 diabetes. Diabetes.
2014;24(12):1967–81. 2011;60(1):315–9.
64. Pruzin JJ, Nelson PT, Abner EL, Arvanitakis 78. de Bresser J, Tiehuis AM, van den Berg E, et  al.
Z.  Invited review: relationship of type 2 diabe- Progression of cerebral atrophy and white matter
tes to human brain pathology. Neuropathol Appl hyperintensities in patients with type 2 diabetes.
Neurobiol. 2018;44(4):347–62. Diabetes Care. 2010;33(6):1309–14.
65. Blevins BL, Vinters HV, Love S, et  al. Brain arte- 79. Wisse LE, de Bresser J, Geerlings MI, et al. Global
riolosclerosis. Acta Neuropathol. 2021;141(1):1–24. brain atrophy but not hippocampal atrophy is related
66. Gerstein HC, Smith EE, Ramasundarahettige to type 2 diabetes. J Neurol Sci. 2014;344(1–2):32–6.
C, et  al. Diabetes, brain infarcts, cognition, and 80. Espeland MA, Bryan RN, Goveas JS, et al. Influence
small vessels in the Canadian Alliance for Healthy of type 2 diabetes on brain volumes and changes in
Hearts and Minds study. J Clin Endocrinol Metab. brain volumes: results from the Women’s Health
2021;106(2):e891–e8. Initiative Magnetic Resonance Imaging studies.
67. Moran C, Beare R, Phan T, et al. Neuroimaging and Diabetes Care. 2013;36(1):90–7.
its relevance to understanding pathways linking dia- 81. Callisaya ML, Beare R, Moran C, Phan T, Wang W,
betes and cognitive dysfunction. J Alzheimers Dis. Srikanth VK. Type 2 diabetes mellitus, brain atrophy
2017;59(2):405–19. and cognitive decline in older people: a longitudinal
68. Traylor M, Persyn E, Tomppo L, et al. Genetic basis study. Diabetologia. 2019;62(3):448–58.
of lacunar stroke: a pooled analysis of individual 82. Groeneveld O, Reijmer Y, Heinen R, et  al. Brain
patient data and genome-wide association studies. imaging correlates of mild cognitive impair-
Lancet Neurol. 2021;20(5):351–61. ment and early dementia in patients with type 2
69. Garfield V, Farmaki AE, Fatemifar G, et  al. The diabetes mellitus. Nutr Metab Cardiovasc Dis.
relationship between glycaemia, cognitive function, 2018;28(12):1253–60.
structural brain outcomes and dementia: a Mendelian 83. Reijmer YD, Brundel M, de Bresser J, et  al.
Randomisation Study in the UK Biobank. Diabetes. Microstructural white matter abnormalities and
2021;70(10):2313–21. cognitive functioning in type 2 diabetes: a dif-
70. Schneider ALC, Selvin E, Sharrett AR, et al. Diabetes, fusion tensor imaging study. Diabetes Care.
prediabetes, and brain volumes and subclinical cere- 2013;36(1):137–44.
Brain Changes in Diabetes and Cognitive Dysfunction 395

84. Sanjari Moghaddam H, Ghazi Sherbaf F, Aarabi 99. American Diabetes Association. 12. Older adults:
MH.  Brain microstructural abnormalities in type 2 standards of medical care in diabetes-2019. Diabetes
diabetes mellitus: a systematic review of diffusion Care. 2019;42(Suppl 1):S139–S47.
tensor imaging studies. Front Neuroendocrinol. 100. Committee Report: Glycemic targets for elderly
2019;55:100782. patients with diabetes: Japan Diabetes Society (JDS)/
85. Alotaibi A, Tench C, Stevenson R, et al. Investigating Japan Geriatrics Society (JGS) Joint Committee on
brain microstructural alterations in type 1 and type 2 Improving Care for Elderly Patients with Diabetes. J
diabetes using diffusion tensor imaging: a system- Diabetes Investig. 2017;8(1):126–8.
atic review. Brain Sci. 2021;11(2):140. 101. Sinclair AJ, Hillson R, Bayer AJ.  Diabetes and
86. den Haan J, Verbraak FD, Visser PJ, Bouwman dementia in older people: a Best Clinical Practice
FH.  Retinal thickness in Alzheimer’s disease: a Statement by a multidisciplinary National Expert
systematic review and meta-analysis. Alzheimers Working Group. Diabet Med. 2014;31(9):1024–31.
Dement (Amst). 2017;6:162–70. 102. LeRoith D, Biessels GJ, Braithwaite SS, et  al.
87. Khalil M, Teunissen CE, Otto M, et  al. Treatment of diabetes in older adults: an Endocrine
Neurofilaments as biomarkers in neurological disor- Society* clinical practice guideline. J Clin
ders. Nat Rev Neurol. 2018;14(10):577–89. Endocrinol Metab. 2019;104(5):1520–74.
88. Bridel C, van Wieringen WN, Zetterberg H, et  al. 103. Brown AF, Mangione CM, Saliba D, Sarkisian
Diagnostic value of cerebrospinal fluid neuro- CA. Guidelines for improving the care of the older
filament light protein in neurology: a system- person with diabetes mellitus. J Am Geriatr Soc.
atic review and meta-analysis. JAMA Neurol. 2003;51(5 Suppl Guidelines):S265–S80.
2019;76(9):1035–48. 104. Srikanth V, Sinclair AJ, Hill-Briggs F, Moran
89. van Golen LW, Huisman MC, Ijzerman RG, et  al. C, Biessels GJ.  Type 2 diabetes and cognitive
Cerebral blood flow and glucose metabolism mea- dysfunction-towards effective management of
sured with positron emission tomography are both comorbidities. Lancet Diabetes Endocrinol.
decreased in human type 1 diabetes. Diabetes. 2020;8(6):535–45.
2013;62(8):2898–904. 105. Smith EE, Saposnik G, Biessels GJ, et  al.
90. Mankovsky BN, Metzger BE, Molitch ME, Prevention of stroke in patients with silent cere-
Biller J.  Cerebrovascular disorders in patients brovascular disease: a scientific statement for
with diabetes mellitus. J Diabetes Complications. Healthcare Professionals From the American Heart
1997;10:228–42. Association/American Stroke Association. Stroke.
91. Jansen JF, van Bussel FC, van de Haar HJ, et  al. 2017;48(2):e44–71.
Cerebral blood flow, blood supply, and cognition in 106. Areosa Sastre A, Vernooij RW, Gonzalez-Colaco
type 2 diabetes mellitus. Sci Rep. 2016;6(1):10. Harmand M, Martinez G. Effect of the treatment of
92. Cui Y, Liang X, Gu H, et al. Cerebral perfusion alter- type 2 diabetes mellitus on the development of cog-
ations in type 2 diabetes and its relation to insulin nitive impairment and dementia. Cochrane Database
resistance and cognitive dysfunction. Brain Imaging Syst Rev. 2017;6:CD003804.
Behav. 2017;11(5):1248–57. 107. Patrone C, Eriksson O, Lindholm D. Diabetes drugs
93. Bangen KJ, Werhane ML, Weigand AJ, et  al. and neurological disorders: new views and thera-
Reduced regional cerebral blood flow relates to peutic possibilities. Lancet Diabetes Endocrinol.
poorer cognition in older adults with type 2 diabetes. 2014;2(3):256–62.
Front Aging Neurosci. 2018;10:270. 108. McMillan JM, Mele BS, Hogan DB, Leung
94. Roberts RO, Knopman DS, Cha RH, et al. Diabetes AA.  Impact of pharmacological treatment of dia-
and elevated hemoglobin A1c levels are associated betes mellitus on dementia risk: systematic review
with brain hypometabolism but not amyloid accu- and meta-analysis. BMJ Open Diabetes Res Care.
mulation. J Nucl Med. 2014;55(5):759–64. 2018;6(1):e000563.
95. Spauwen PJ, van Eupen MG, Kohler S, et  al. 109. Biessels GJ, Verhagen C, Janssen J, et al. Effects of
Associations of advanced glycation end-products linagliptin vs glimepiride on cognitive performance
with cognitive functions in individuals with and in type 2 diabetes: results of the randomised double-­
without type 2 diabetes: the maastricht study. J Clin blind, active-controlled CAROLINA-COGNITION
Endocrinol Metab. 2015;100(3):951–60. study. Diabetologia. 2021;64(6):1235–45.
96. Moran C, Munch G, Forbes JM, et  al. Type 2 dia- 110. Biessels GJ, Verhagen C, Janssen J, et  al. Effect
betes, skin autofluorescence, and brain atrophy. of linagliptin on cognitive performance in patients
Diabetes. 2015;64(1):279–83. with type 2 diabetes and cardiorenal comorbidities:
97. Biessels GJ, Whitmer RA.  Cognitive dysfunction the CARMELINA randomized trial. Diabetes Care.
in diabetes: how to implement emerging guidelines. 2019;42(10):1930–8.
Diabetologia. 2020;63(1):3–9. 111. Gerstein HC, Hart R, Colhoun HM, et al. The effect
98. Federation ID.  Global guideline for managing of dulaglutide on stroke: an exploratory analysis
older people with type 2 diabetes. 2013. https:// of the REWIND trial. Lancet Diabetes Endocrinol.
www.idf.org/e-­l ibrary/guidelines/78-­g lobal-­ 2020;8(2):106–14.
guideline-­f or-­m anaging-­o lder-­p eople-­w ith-­t ype-­ 112. Kirkman MS, Briscoe VJ, Clark N, et al. Diabetes in
2-­diabetes.html. older adults. Diabetes Care. 2012;35(12):2650–64.
Lifestyle and Dietary
Modifications: Relevance
in the Management of Diabetic
Neuropathy

Jonathan Enders and Douglas E. Wright

1 Introduction could lead to the identification of new drug


targets.
Diabetic peripheral neuropathy (DPN) is a com-
mon complication of prediabetes, type 1 diabetes
mellitus (T1DM), and type 2 diabetes mellitus 2 Pharmacologic Treatment
(T2DM). DPN remains largely irreversible, and of Diabetic Peripheral
there are no known curative treatments for Neuropathy
patients with DPN [1, 2]. The discovery of preci-
sion therapeutics remains an important goal since For many patients with T2DM, the biguanide
a clear understanding of the breadth of pathogen- metformin is commonly recommended as first
esis remains elusive. It is known that strict glyce- line therapy following lifestyle modifications.
mic control using oral agents or insulin therapy However, other medications—including insulin
can slow or prevent the progression of DPN [3, injection, alpha-glucosidase inhibitors, dipepti-
4]. Worldwide, the incidence of diabetes and obe- dyl peptidase-4 (DPP-4) inhibitors, glucagon-­
sity is linked and increasing dramatically, helping like peptide-1 receptor agonists, sodium-glucose
to drive an increase in the prevalence of transporter (SGLT) 2 inhibitors—can also be
DPN.  Driven by axon degeneration, demyelin- effective by themselves or in combination ther-
ation, and nerve dysfunction, common symptoms apy [6]. Glycemic control in patients with T1DM
of DPN include loss of sensation, limb pain, gait is generally addressed with insulin injections [7].
disturbance, and autonomic dysfunction [5]. Achieving tight glycemic control is challenging,
Collectively, these symptoms reduce the quality as meaningful reductions in blood glucose is the
of life for patients with DPN.  This chapter will desired goal, but this outcome is strongly influ-
highlight experimental approaches to explore enced by patient compliance, activity level, diet,
dietary interventions and lifestyle changes to and overall health.
treat DPN.  The chapter will also address their For pain associated with DPN, there are sev-
limitations and discuss how these interventions eral pharmacologic therapies used with variable
success. A subset (10–26%) of patients with DPN
report pain that is described as stabbing, electri-
J. Enders ∙ D. E. Wright (*) cal pain, pins and needles, or burning sensations
Department of Anesthesiology, University of Kansas [8]. It is thought that tight glycemic control is not
Medical Center, Kansas City, KS, USA sufficient to completely prevent painful symp-
e-mail: dwright@kumc.edu toms [9, 10]. Pain medications include tricyclic

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 397
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_22
398 J. Enders and D. E. Wright

antidepressants (TCAs), serotonin and below in this chapter should all consider obesity
­norepinephrine reuptake inhibitors (SNRIs), and as a key factor in DPN improvements.
anticonvulsants (gabapentin or pregabalin) [11,
12]. As an over-the-counter supplement, α-lipoic
acid has shown moderate efficacy in studies of 4 High-Fat Diets as a Tool
DPN-­associated pain [13, 14]. Finally, topical to Study Diabetic Peripheral
treatments can be used that includes application Neuropathy
of a lidocaine patch or capsaicin cream [15].
There are also other off-label medications used to A healthy diet provides clear benefit to prevent-
treat painful DPN, including herbal supplements ing diabetic neuropathy and using a high-fat diet
like nutmeg and Citrullus colocynthis extract [16, (HFD) in the setting of DPN has revealed impor-
17]. Opioids generally are contraindicated for tant insights into mechanisms and outcomes
DPN-associated pain. Overall, these classes of associated with a poor diet. Poor diet is risk fac-
medications are not sufficiently effective and tor for the development of T2DM but has the
commonly have physiological or psychological potential to be modified. Using a high-fat diet in
side effects that limit their use. Thus, additional animal models has revealed extensive informa-
treatments to clinically treat DPN are needed. tion about how elevated carbohydrates and fats
Hopefully, when coupled with lifestyle or dietary can lead to symptoms that strongly resemble pre-
changes, the effectiveness of these existing diabetes and, if carried out long enough, manifes-
approaches may be improved. tations of diabetes and neuropathy. This has been
covered extensively in a number of reviews [19].
One advantage of using dietary interventions is
3 Obesity and Diabetic that neuropathy progression is more gradual and
Peripheral Neuropathy leads to metabolic imbalances resembling the
natural progression from obesity to T2DM.  In
It is important to understand the role of obesity in addition, HFDs also provide stronger relevance
DPN, as obesity is increasing rapidly and is rele- to increased dietary fat/dyslipidemia in both met-
vant to lifestyle and dietary modifications that abolic syndrome in patients with prediabetes and
impact DPN.  Both preclinical and clinical T2DM patients [20–23].
research have clearly identified obesity as a key In general terms, chronic use of experimental
risk factor for DPN.  Recent work by Callahan HFD paradigms can lead to progressive impair-
and colleagues has shown in studies from various ments in sensori-behavioral changes, nerve con-
countries that obesity strongly associates with duction, and nerve pathology. These dietary
DPN [11]. Other metabolic syndrome compo- formulations generally range from 25% fat/40%
nents, such as hypertriglyceridemia, hyperten- carbohydrates up to 60%fat/20% carbohydrates
sion, and low high-density lipoprotein cholesterol, [19] (24% from fat, 41% from carbohydrates).
appear less strongly commonly associated with Common complications in HFD-fed mice include
DPN compared with obesity and hyperglycemia. weight gain and glucose intolerance, and as well
Importantly normoglycemic patients with obe- as impairments in aforementioned neurological
sity have a higher prevalence of DPN than lean, impairments that include, including behavioral
healthy patients, strongly suggesting obesity (hindpaw thermal and mechanical threshold
itself is an independent risk factor of DSP [11]. changes), slowed nerve conduction velocity, and
This point should strongly influence preclinical sensory nerve terminal degeneration (intraepider-
studies (discussed below) that use dietary inter- mal nerve fiber density: IENFD), leading to
ventions to model DPN. Weight loss and exercise reduced innervation of the cutaneous tissue [19].
are still believed to be one of the best treatments Some of the potential mechanisms identified that
for patients that suffer from obesity-related DPN contribute to these deficits include weight gain,
[18]. The lifestyle and dietary changes described insulin resistance of sensory ­neurons [24, 25],
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 399

dyslipidemia [20], mitochondrial deficits [26], tion [38] and poor mitochondrial trafficking/
and growth factor changes [27]. health [39–41]. Relevant to DPN, ROS can also
In addition to mice, adult Sprague-Dawley impact several transient receptor potential (TRP)
(SD) rats have been studied extensively when channels [42–45] and also sensitize voltage-gated
exposed to a HFD.  These studies have largely channels [46, 47] to evoke abnormal sensory sig-
revealed similar findings that include abnormal naling that is evident in DPN.
changes that include the appearance of mechani-
cal allodynia, reduced thermal nociception as
decreased IENFD and changes in subepithelial 5 Dietary Interventions
corneal nerves [28]. Another rat model that to Improve Diabetic
experimentally highlights metabolic complica- Peripheral Neuropathy
tions includes rats selectively bred for high (high-­
capacity runners; HCR) or low endurance 5.1 Mediterranean Diets
exercise capacity (low-capacity runners; LCR)
[29]. Selective breeding of these rats has resulted Although relatively understudied, a dietary inter-
in divergent intrinsic aerobic capacities and sus- vention gaining preclinical interest in DPN is the
ceptibility for metabolic complications. Analysis Mediterranean diet. The Mediterranean diet is
of these rats revealed that LCR female rats had less stringent compared to other diets in terms of
higher overall fat content, surprisingly higher macronutrient abundance. Namely, the
IENFD, reduced mechanical sensitivity, and Mediterranean diet is rich in whole grains, vege-
increased numbers of mast and Langerhans cells tables, and legumes, with olive oils as a primary
in skin compared to female HCR rats. These source of fats and poultry and fish as the main
results suggest that poor metabolic status and source of protein. As a result, adherence to a
reduced aerobic capacity influence peripheral Mediterranean diet results in reduced sucrose
sensation and inflammation in peripheral tissues intake and elevated consumption of monounsatu-
that are involved in DPN [29]. Continued study rated and omega-3 polyunsaturated fatty acids.
of the HCR and LCR rats should prove fruitful to One potential benefit of the Mediterranean
understand how metabolic complications impact diet for treating DPN is relieving hyperglycemia.
sensory function and pain. Finally, sex differ- Two studies from Esposito and colleagues sug-
ences in the quality and sensitivity of male and gest that patients with T2DM consuming a
female rats have been reported [30], and potential Mediterranean diet were less likely to experience
sex differences likely require increased rigor and hyperglycemia and exhibit reduced hemoglobin
attention. Overall, the use of dietary intervention A1C.  These benefits may in part be due to
and genetic breeding to model metabolism asso- improved insulin sensitivity [48, 49]. With whole
ciated DPN features has provided unique insight grain as the primary carbohydrate source of
to the role of obesity, insulin resistance, dyslipid- Mediterranean diets, sucrose and fructose intake
emia, inflammation in DPN. are reduced, which has been linked preclinically
Diet is critical in modifying the redox state of to spared insulin resistance and rescued neuro-
sensory neurons and, thus, the progression of pathic symptoms such as mechanical allodynia
DPN. Consumption of a HFD leads to hypergly- and intraepidermal fiber loss [50, 51].
cemia and dyslipidemia. Hyperglycemia results Mediterranean diets may also offer benefit in
in the accumulation of toxic metabolites, such as DPN by reversing dyslipidemia. Feldman and
sorbitol [31, 32] and reactive dicarbonyls that colleagues have demonstrated many harmful
include methylglyoxal and glyoxal [33]. These effects of dyslipidemia in DPN, such as poor
toxic metabolites increase the generation of [34] mitochondrial trafficking and health, that contrib-
and impair the scavenging of ROS [35–37]. ute to disease progression [39, 40]. Preclinical
HFD-induced dyslipidemia is also a key risk fac- studies by Rumora et  al. reveal that mice fed a
tor for DPN, resulting in elevated ROS genera- HFD composed primarily of saturated fatty acids
400 J. Enders and D. E. Wright

go on to develop symptoms of peripheral neurop- about the efficacy of Mediterranean diets as inter-
athy, while mice fed a diet high in monounsatu- ventions for DPN.
rated fats, like those found in olive oil, did not
develop neuropathy [52]. Moreover, monounsatu-
rated fat-rich diets reversed the neuropathy caused 5.2 Ketogenic Diets
by high saturated fat diets in these animals and
were able to rescue mitochondrial motility and Ketogenic diets are very low carbohydrates that
health [52]. Thus, preclinical models that resem- lead to ketogenesis by the liver. Ketogenic diets
ble a Mediterranean diet prevent and reverse the have been used to treat neurodegenerative disor-
onset of DPN and obesity-related neuropathies. ders [54, 55], epilepsy and seizures [56, 57], and
While there is accumulating preclinical evi- chronic pain [58–60]. The outcome of a keto-
dence that suggests potential efficacy of a genic diet is to displace the carbohydrate with
Mediterranean diet in treating DPN, to the best of oils and fats, and the resulting effect of excess fat
our knowledge no explicit clinical study of consumption causes increased liver fatty acid
Mediterranean diet in DPN or other neuropathic oxidation and production of the two major ketone
conditions has been completed. There is, how- bodies β-hydroxybutyrate (β-OHB) and acetoac-
ever, limited evidence that increased omega-3 etate. Once synthesized, these two ketone bodies
fatty acid consumption is negatively correlated enter circulation and are taken up by extra-hepatic
with incidence of DPN [53], again supporting a tissues to replace glucose as a principal fuel
putative protective effect for Mediterranean diets. source.
There remains, however, a significant gap in Because ketogenic diets reduce glucose intake
research that precludes a definitive statement and circulating levels of glucose, ketogenic diets

Fig. 1 Systemic
changes induced by a
ketogenic diet. A Reduced
ketogenic diet induces Hyperglycemia
widespread benefits to
many organ systems
involved in
DPN. Reduced
consumption of Detoxify Toxic
carbohydrates due to Metabolites
consuming a ketogenic
diet reduces
hyperglycemia and
accumulation of
advanced glycation Nutritional Reduced
endproducts. Moreover,
Ketosis Inflammation
ketogenic diets
contribute to
detoxification of toxic
metabolites, reduced
inflammation and Reduced
generation of reactive Oxidative Stress
oxygen species, and
support peripheral axon
growth

Support
Peripheral
Axon Growth
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 401

have been used in the treatment of patients with their safety in patients with diabetes. These clini-
diabetes (Fig. 1). Prior to the widespread use of cal studies are strongly consistent with findings
insulin treatment, low-carbohydrate diets were in preclinical models of prediabetes [58], T1DM
commonly used to reduce elevated glucose levels and T2DM [67–69]. Although more investigation
through dietary reduction of carbohydrates in is needed, there is increasing and powerful evi-
mild cases of diabetes and more severe elimina- dence that ketogenic diets can be safely used in
tion of protein and carbohydrate consumption in patients with DPN.
severe cases of diabetes [61]. Patients with diabe-
tes that were fed low-carbohydrate meals did not 5.2.2 Ketogenic Diet and Neural
exhibit post-prandial hyperglycemia similar to Disease
diabetic patients consuming a standard diet [62]. Many of the benefits of using ketogenic diets to
Importantly, these diets did not originally replace treat neural disorders were grounded in their use
lost caloric intake from reduced carbohydrate for the treatment of pharmacologically non-­
consumption with elevated fat intake, leading to responsive epilepsy. Now, ketogenic diets have
severe morbidity. Complications of this starva- been shown to be effective in treating a range of
tion approach often included patient weakness neurodegenerative diseases and traumatic injury
and death. In the early 1920s, it became more [22, 35, 70–73]. Despite their use, however,
common to use high-fat, low-carbohydrate diets there remains a limited understanding of the
in weak and starvation-resistant diabetics [63]. mechanisms of action provided by a ketogenic
These early low-carbohydrate diets resemble diet on chronic painful conditions and
modern ketogenic diets, and their use was suc- DPN.  Regarding various pain conditions we
cessful in improving diabetes parameters in these reported that consumption of a ketogenic diet
patients (Fig. 1). However, following the discov- abruptly improved abnormal mechanical sensa-
ery of and practice of using insulin, emphasis on tion without substantial changes in weight using
high-fat, low-carbohydrate dietary interventions a HFD mouse model [58–60, 67]. In addition,
for diabetes became less common. we have recently demonstrated significant bene-
fits of using a ketogenic diet in reversing
5.2.1 Addressing Health Concerns mechanical allodynia and abnormal thermal sen-
of Using a Ketogenic Diet sation in a mouse model of T1DM [67]. Both of
in Patients with Diabetes these studies also demonstrated, in  vitro, that
Consumption of low-carbohydrate/high-fat diets administration of β-OHD enhanced neurite out-
generates appropriate health concerns, as there is growth from cultured primary sensory neurons
the potential to cause diabetic ketoacidosis, a [58]. Also, both studies also demonstrated,
common life-threatening complication of dia- in  vivo, that consumption of a ketogenic diet
betes. Diabetic ketoacidosis is associated with could prevent reductions in IENFD and, impor-
an accumulation of excess blood ketone bodies tantly, stimulate epidermal nerve fiber regenera-
and lactate. β-OHB, acetoacetate, and lactate tion (Fig.  1) [67]. These studies hold promise
deprotonate at physiologic pH, causing reduc- that using ketogenic diets to treat DPN can pro-
tions in blood pH. With a reduction in carbohy- vide benefits, yet mechanisms by which the diet
drate consumption, however, lactate production or ketone bodies directly or indirectly reduce
is actually decreased [64], thus physiologically nociception and normalize epidermal fiber inner-
sparing patients from acidosis. In fact, acidosis vation are not clearly understood. However, data
was reversed in patients with diabetes consum- from other studies described below point to plau-
ing very low-­carbohydrate, high-fat diets [63]. sible mechanisms that should be explored and
Additional studies in humans reveal [57, 65, 66] will likely provide insight into potential mecha-
the benefits of ketogenic diets in diabetes and nisms and identify new drug targets.
402 J. Enders and D. E. Wright

5.2.3 Ketone Bodies as Signaling regeneration induced by dimethyl fumarate is


Molecules linked to a shift in CD4+ T cell activation toward
Ketone bodies serve as important bioenergetic a TH2 subtype [70]. This is interesting since
fuel sources; yet they are also very effective β-OHB is known to inhibit the NOD- LRR- and
extra-hepatic signaling molecules [74]. It is Pyrin domain-containing protein 3 (NLRP3)
becoming clear that these signaling functions are inflammasome [73, 76]. NLRP3 activation
involved modulating peripheral tissues These cleaves pro-interleukin-1 to interleukin-1β (IL-­
include activation of G-protein coupled receptor 1β), which promotes T cell polarization toward a
signaling cascades, modulation of cellular TH1 phenotype [77, 78] and pro-nociceptive
inflammation, neurotrophin signaling, and epi- responses [79, 80]. Together, these studies pro-
genetic modifications. β-OHB serves an extracel- vide support that ketone bodies may improve
lular ligand for hydroxycarboxylic acid receptor sensory neuron function by modulating inflam-
2 (HCAR2) [75], a G-protein coupled receptor mation, a key risk factor in DPN (Figs. 1 and 2).
widely expressed in a range of tissues, including An additional signaling function of β-OHB is
DPN-relevant nociceptors and circulating related to its activity as a histone deacetylase
immune cells (Fig.  2). Dimethyl fumarate is (HDAC) inhibitor. Exercised-induced elevations
another HCAR2 ligand and has shown promise in of β-OHB are associated with increased skeletal
preclinical pain models [75] and axonal degen- muscle expression of brain-derived neurotrophic
eration following nerve injury [70]. Axonal factor (BDNF) [81]. β-OHB increased activity at

Inflammation

Nociceptive b-HB
Priming
K
HCAR2 K-ATP
MCT1 K
Channels K

αi β/y
ATP
IL-1b b-HB

Glycolysis
NRF2
Membrane
Associated
ATP

Cytosolic ATP
NLRP3
ROS
lnflammasome HDACs Mitochondrial
Biogenesis &
Antioxidant Mass
Genes Bdnf

Fig. 2 Molecular functions of β-hydroxybutyrate NLRP3 inflammasome, activation of NRF2-mediated


(β-OHB). β-OHB mediates its effects by extracellular sig- transcriptional profiles, inhibition of histone deacetylases,
naling through G-protein-coupled receptors like HCAR2 improved mitochondrial biogenesis and function, and
and through intracellular interactions after entering the reduced glycolysis, resulting in activation of KATP
cell through the monocarboxylic acid transporter 1 channels
(MCT1). These effects include reduced activation of the
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 403

the BDNF promoter and decreased HDAC activ- Heightened nociception caused by toxic gly-
ity and occupancy of the BDNF promoter by colysis metabolites and ROS may be potentially
HDAC2 and HDAC3. β-OHB also rescued modulated by a ketogenic diet or ketone adminis-
HDAC hyperactivity in settings of neuropathic tration. The antioxidant effects of a ketogenic diet
pain, spinal cord injury [72, 82], and cisplatin-­ are well established [93]. As an example relevant
induced neuropathy [83]. Thus, β-OHB modula- to DPN, we reported that a ketogenic diet reduces
tion of the epigenetic landscape via HDAC H2O2 generation in the sciatic nerve [94] of dia-
inhibition provides an additional mechanism that betic mice, this effect was seen in the distal nerve
merits exploration (Fig. 2). segment only, suggesting a length-­dependent pro-
In addition to β-OHB, acetoacetate may also cess that could be associated with impaired mito-
serve important functions associated with chondrial trafficking in DPN [94]. Finally,
DPN.  In a metabolism-independent fashion, methylglyoxal synthesis occurs as a normal con-
acetoacetate is known to stimulate extracellular-­ sequence of glycolysis; thus, methylglyoxal accu-
signal related kinase (ERK) activation [23]. mulation should be reduced during ketosis.
Treatment with acetoacetate increased prolifer- Recent evidence now suggests that a ketogenic
ation of skeletal muscle satellite cells [84] that diet may enhance axon degeneration and regen-
was associated with increased entry into S eration in DPN. DPN leads to reductions in epi-
phase and phosphorylation of the kinases ERK1 dermal nerve fibers [95–98] and demyelination of
and ERK2. These effects occurred in the pres- large-diameter axons [99, 100]. Hyperglycemia
ence of knockdown of Oxct1, the enzyme contributes to this neurodegenerative change in
responsible for catalyzing the generation of peripheral innervation, and Schwann cells cul-
acetyl-CoA from acetoacetate, suggesting the tured in high glucose conditions undergo apopto-
effects were independent of acetoacetate sis [101] and de-­ differentiation [102].
metabolism [85]. Additionally, the Schwann cells secrete less NGF,
leading to reduced DRG neurite outgrowth [103].
5.2.4 Ketogenic Diets and TRP We recently published that consumption of a
Channels ketogenic diet stimulates the regeneration of epi-
The modulation of TRP ion channels is a poten- dermal nerve fibers in a mouse model of DPN
tial mechanism to explain the actions of keto- [67]. The increased axon regeneration is likely
genic diets. TRP ankyrin 1 (TRPA1) and TRP associated with both improved glycemic status
vanilloid 1 (TRPV1) are members of the transient and direct actions of ketone bodies on DRG neu-
receptor potential family of ion channels and are rons. This latter point is based on in vitro studies,
strongly implicated in painful conditions, includ- where β-OHB stimulated neurite outgrowth in
ing painful DPN [6, 31, 86–89]. These channels cultured DRG neurons in both the presence and
are responsive to toxic metabolites known to be absence of glucose [58]. This in vitro work sug-
upregulated in diabetes, such as methylglyoxal gests that ketone bodies have direct actions to
[90] and ROS [42–44]. Capsaicin sensitivity of stimulate axon growth, perhaps through serving
TRPV1 is heightened in DRG neurons in diabetic as a potent fuel source (Fig. 1) [58].
rats [91]. The heightened activity can be rescued
by supplementation of the antioxidant melatonin, 5.2.5 Ketones and Modulation
implicating ROS and ROS-mediated TRPVI sen- of ATP-Gated Ion Channels
sitization in DPN.  Methylglyoxal can activate Ketogenic diets influence neuronal excitability
TRPA1 to induce abnormal neuronal activity and via regulation of ATP-gated potassium (KATP)
nociception [92]. Elevated ROS in DRG neurons channels, and this action is believed to underlie
[43] and spinal dorsal horn [44] both increase benefits of ketogenic diets in modulation of neu-
spontaneous neuronal activity that can be ronal firing rates in models of seizure and epi-
reversed with TRPA1 and TRPV1 antagonists lepsy (Fig. 2). In the presence of ATP, these ion
[43, 44] (Fig. 1). channels remain closed. There are several pro-
404 J. Enders and D. E. Wright

posed mechanisms thought to be involved in dynia following nerve injury [111].


mechanisms through which a ketogenic diet Glibenclamide use also increases hindpaw
modulates KATP channel activity. Both β-OHB mechanical sensitivity in a model of bone
and acetoacetate reduce the firing rates of cancer-­associated pain in rats [112].
GABAergic neurons in the substantia nigra pars Interestingly, glibenclamide, also known as gly-
reticula [88]. Importantly, this effect occurs in the buride, is an available but infrequently used
presence of glucose, and the inhibitory effect of medication used to treat T2DM [113].
the ketones could be reversed following ketone Specifically related to DPN, a role for KATP chan-
washout, administration of the KATP antagonist nels has not yet been identified. However, the
tolbutamide, or genetic knockout of the Kir6.2 expression of KATP channels in sensory neurons
channel subunit. Additional support is provided and the consequences of hyperglycemia and dys-
from studies of hippocampal slices, where reduc- lipidemia on channel activity suggest KATP chan-
tions in available glucose lead to hyperpolariza- nels may be an important cellular target. It is
tion of CA3 pyramidal neurons and decreased known that KATP channel opening is affected by
frequencies of post-synaptic currents [104], The glucose concentration [114, 115], and
KATP antagonist tolbutamide similarly prevented hyperglycemia-­ evoked neuronal depolarization
and reversed outward potassium currents in these of DRG neurons can be reversed by diazoxide, a
neurons in reduced glucose. KATP channel activator [116]. As a surrogate for
Through the reduction of carbohydrate intake, dyslipidemia, palmitate supplementation lowers
evidence suggests that a ketogenic diet reduces the membrane-localization of the KATP channel
glycolysis that then leads to reductions in plasma subunit SUR1, resulting in cellular lipotoxicity
membrane-proximal ATP. The result of this effect [117]. Collectively, the effect of ketogenic diets
would then increase the opening of KATP chan- and ketone bodies to reduce glucose utilization,
nels. Because glycolysis is partially localized to glycolytic flux, and hyperglycemia [69, 107,
the plasma membrane, it can provide a localized 108] could increase opening of KATP channels
source of ATP adjacent to cellular pumps that and ultimately reduce neuronal firing in neurons
includes the Na+/K+-ATPase pump [105, 106]. [88, 104]. Also, ketogenic diets also reduce dys-
Ketone bodies are known to reduce cellular glu- lipidemia [85, 118]. Based on this growing lit-
cose utilization and glycolytic flux [107, 108], erature, it is plausible that a ketogenic diet could
which reduces ATP gradients and increases KATP function in an antinociceptive action in DPN and
channel activity. Similarly, reducing total glu- KATP channels may be an important target to
cose, in essence, decreases substrate availability, explore.
leading to decreased ATP and decreased opening
of KATP channels. 5.2.6 Exogenous Ketone
Importantly, KATP channels have been Supplementation as a Tool
reported to play a role in modulation of pain and to Treat Diabetic Peripheral
nociception. KATP channels are expressed by Neuropathy
sensory neurons [109], and opening of KATP Evidence is growing that ketogenic diets may
channels reduces neuronal current amplitude serve as a promising intervention for treating
following electrical stimulus. Inhibiting KATP DPN and other chronic pain conditions. However,
channels with the antagonist glibenclamide long term adherence to a ketogenic diet remains
exerts an opposite effect to increase current difficult and may ultimately be difficult for
amplitude. Ligation of the sciatic nerve closes patients to follow. This limitation may be over-
KATP channels in sensory neurons [110] and come by direct supplement with exogenous
leads to a decrease in KATP channel expression in ketone bodies. There is a growing market that
the spinal cord [109, 111]. Conversely, intrathe- promotes commercially available ketone salts,
cal administration of a KATP channel activator ketone salts with medium-chain triglyceride
reverses thermal algesia and mechanical allo- (MCT) oil, or ketone esters [91, 99]. At this point,
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 405

there is no clinical evidence regarding ketone One mechanism by which alterations in


supplementation in DPN.  However, evidence microbiota may contribute to the onset of neuro-
suggests that exogenous ketone supplementation pathic pain is through dysregulation of pathogen-
mirrors some of the positive actions of a keto- or damage-associated molecular pattern
genic diet. Addition a β-OHB salt with MCT oil receptors, such as the toll-like receptor (TLR)
or ketone ester can reduce blood glucose [119], family (Fig. 3). These receptors play an essential
and supplementation with ketone salts reduces role in innate immunity by detecting molecular
oxidative stress in select organ systems [120]. It motifs normally associated with infection and
is likely that certain actions (axon growth, initiating an inflammatory response. TLR4, for
G-protein receptor activation, epigenetic regula- instance, is the canonical receptor for lipopoly-
tion, ion channel modulation) of a ketogenic diet saccharide (LPS) and, when activated, drives
on DPN may be independent of hyperglycemia symptoms of neuropathy in healthy mice [123]
and could be mimicked through ketone supple- and contributes to onset and progression of
mentation. This will be an important area for obesity-­induced [124, 125] and chemotherapy-­
research as it may be an easier intervention for induced neuropathy [126].
patients with diabetes and can be easily coupled Another potential mechanism includes signal-
with existing medications. ing by metabolites produced by microbiota.
Butyrate, for instance, is commonly produced as a
metabolic byproduct and signaling metabolite in
6 Dietary Influences gut flora [127]. Butyrate is a potent inhibitor of his-
on the Microbiome Relevant tone deacetylases (HDACs) [128, 129] and may
to Diabetic Peripheral play a similar role to the closely related
Neuropathy β-hydroxybutyrate in regulating gene expression of
neurotrophins [81]. Butyrate is directly correlated
Another mechanism by which lifestyle interven- with improvements in allodynia and intraepidermal
tion might affect the severity of DPN is through nerve density in obese mice following fecal trans-
regulation of gut microbiota. The microbiome is fer [71]. Interestingly, butyrate also demonstrates
emerging as a novel perspective in several pathol- activity as a partial agonist of the capsaicin recep-
ogies, and its involvement in pain and neuropathy tor, transient receptor channel vanilloid 1 (TRPV1),
is emerging. Mansuy-Aubert recently used a and increases desensitization of TRPV1 [71].
moderately high-fat Western diet to induce neu- TRPV1 contributes to DPN and other neuropathic
ropathy in mice. They then assessed differences conditions [87, 91, 95, 97, 130]; thus, its modula-
in microbiota populations and performed fecal tion by butyrate, and presumably butyrate-produc-
transplantation [71]. Mice that were fed a Western ing bacteria, provides an exciting avenue of
diet developed symptoms of neuropathy and, research moving forward (Fig. 3).
importantly, displayed reduced diversity in gut While sedentary lifestyles and Western diet
flora and abundance of butyrate-producing bacte- contribute to changes in microbiota that are pro-­
rial species. Antibiotics and subsequent fecal pathogenic, lifestyle interventions also alter the
transfer rescued mouse thermal and mechanical gut microbiota. As previously discussed, keto-
sensory symptoms, increased intraepidermal genic diets are gaining popularity culturally and
innervation, and induced transcriptional changes as therapeutic interventions for a variety of pain
in the DRG and sciatic nerves. These effects are conditions. There is strong evidence that these
consistent with the effects of antibiotic treatment diets profoundly alter gut microbiota, though
[121] and fecal transfer [122] in reversing these changes have yet to be documented in the
chemotherapy-­ induced neuropathy, suggesting context of diabetes and DPN.  In children with
that alterations in gut microbiota may contribute severe epilepsy, a ketogenic diet decreased
to painful neuropathies generally, not just in dia- Actinobacteria and Eubacterium rectale, a major
betes (Fig. 3). butyrate-producing species [131]. There are con-
406 J. Enders and D. E. Wright

Butyrate Lipopolysaccharide
(LPS)

HDAC TRPV1 TLR4 Signaling


Inhibition Regulation
Altered Gene Reduced
Partial
Expression Inflammation
Agonism
Increased Increased Reduced Nociceptive
Neurotrophins Desensitization Priming

Fig. 3  The effect of the microbiome on DPN. The micro- flora appears to have antinociceptive actions. Butyrate
biota is known to impact neuropathy and play important acts as a partial agonist for the capsaicin receptor TRPV1
putative roles in DPN through varied mechanisms. and increases desensitization of this receptor. Additionally,
Increased systemic lipopolysaccharide (LPS) is pro-­ butyrate acts as a histone deacetylase (HDAC) inhibitor,
nociceptive, activating TLR4 signaling and driving where it contributes to cell survival programs and tran-
inflammation. Conversely, butyrate production by gut scription of neurotrophins

flicting reports whether ketogenic diets decrease related to the onset and progression of DPN. It is
[132] or increase [133] butyrate production in the known that voluntary exercise positively affects
gut, though these differences may be due to adher- hyperglycemia, insulin resistance [134, 135],
ence to the ketogenic diet and the clinical popula- dyslipidemia [86], and advanced glycation
tions being examined. If ketogenic diets indeed ­endproducts (AGE) accumulation [135, 136]. In
reduce microbiota butyrate production, it may be addition IENFD [27, 136] and nerve conduction
that the structurally similar β-hydroxybutyrate velocity (NCV) [137] are altered by exercise
may compensate for reduced butyrate, acting as intervention. Here, we discuss the impact of exer-
an HDAC inhibitor and possibly engaging in cise and physical activity on non-neuronal and
TRPV1 modulation as well. These exciting lines neuronal systems as they relate to DPN.
of research clearly merit more investigation.

7.1 Exercise Benefits on Non-­


7 Exercise and Physical neuronal Tissues
Activity to Improve Diabetic
Peripheral Neuropathy Exercise plays an important role in opposing obe-
sity and metabolic syndrome, major contributors
Exercise and physical activity interventions are to prediabetes and T2DM.  Exercise improves
important and increasingly studied lifestyle inter- body composition, a risk factor relevant to DPN
ventions for DPN. Exercise interventions can be in T2DM [138]. Exercise can improve bone min-
reasonably modeled preclinically and preclinical eral density in premenopausal women and in
studies in rodents have revealed beneficial effects males [89, 139, 140]. Additionally, the Health,
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 407

Aging, and Body Composition study identified a patients with DPN have reported increased
relationship between bone mineral density and IENFD and sprouting in proximal leg skin biop-
sensory threshold, NCV, and nerve conduction sies [136]. Consistent with these findings, a par-
amplitude [141]. Although interesting, relation- allel study using 12 weeks of a similar exercise
ship between reduced bone mineral density and program reported a similar, significant increase
DPN is not clear and requires further study. in distal leg IENFD [149]. Here, clinical research
Dyslipidemia induces mitochondrial damage seems to indicate a positive effect of IENFD and
[39, 40], increased ROS, and inflammation in suggests additional studies are needed.
many tissues. In addition, dyslipidemia and insu- One important and clear effect of exercise is
lin resistance engage in a positive feedback loop improvements in mitochondrial health. The
[142, 143]. Exercise reduces circulating triglyc- extreme morphology and axon length of DRG
eride levels in metabolic syndrome and diabetes axons require a significant energetic demand. It
[86]. Finally, aerobic exercise consistently has been proposed that in DPN, mitochondrial
improves insulin resistance in both preclinical damage disrupts energy production in sensory
studies of diabetes [135] and in clinical settings axons, leading to axon damage and degeneration
[134]. Importantly, exercise reduces insulin resis- [39, 40, 150]. Exercise is known to increase the
tance and hyperglycemia in DPN in T1DM and expression of heat shock protein 70 (HSP70) in
T2DM [135]. Importantly, this leads to reduced the DRG of diabetic rats [151]. HSP70 plays an
AGE accumulation in individuals that exercise essential role in chaperone-mediated autophagy
[136]. mitophagy [152]. Moreover, exercise has been
shown to upregulate peroxisome proliferator acti-
vated receptor gamma associated coactivator 1
7.2 Exercise Benefits on Neuronal alpha (PGC1α), a critical mediator of mitochon-
Tissues drial biogenesis [153, 154]. Overexpression of
downstream targets of PGC1α in sensory neurons
Exercise has direct and demonstrable actions on is sufficient to reverse the painful symptoms and
sensory neurons relevant to DPN.  Exercise is IENFD reductions in mouse models of DPN
known to powerfully regulate neurotrophin [155, 156]. Overall, the evidence for using exer-
expression in tissues innervated by sensory cise as an intervention to improve symptoms of
axons. For example, exercise increases the DPN is exciting. Yet exercise recommendations
expression and release of brain-derived neuro- for T1DM and T2DM patients are not universal
trophic factor (BDNF) [81] and neurotrophin-3 and improved behavioral interventions and edu-
(NT-3) [144] in skeletal muscle. Both neuro- cation for patients with DPN are needed to help
trophins attract proprioceptive axons and are vital them successfully engage in safe, appropriate
to their growth and maintenance. These fibers are exercise regimens.
commonly affected in large-fiber DPN [145–
147], resulting in poor proprioception, altered
gait, and balance control. Preclinical studies have 8 Conclusion
shown that exercise leads to improvements in
these sensory deficits [148] possibly through With the worldwide epidemic increase in obesity
improved muscle spindle innervation, likely and diabetes, impaired quality of life for patients
associated with skeletal muscle neurotrophin that suffer from DPN demands new and better
availability. However, additional studies are nec- non-pharmacological interventions to help slow,
essary to clearly elucidate the connection between reduce or possibly reverse symptoms of
exercise and muscle spindle innervation in DPN. DPN.  Interventions such as dietary changes or
Exercise also impacts IENFD, which is exercise point to important lifestyle changes that
reduced in DPN and often used as a measure of can reduce the burden of DPN, but also highlight
disease progression. Following aerobic and resis- a rich new area to explore novel mechanisms that
tance training for 10  weeks, clinical studies in could lead to the development of new drug
408 J. Enders and D. E. Wright

Obesity
Insulin Resistance
Hyperglycemia
AGE accumulation
Neurotrophins

ROS reduction

Dyslipidemia
Mitochondrial
function

Fig. 4  Points of convergence for dietary- and exercise-­ trophin production, reduced ROS production, and
induced mechanisms. The benefits of dietary intervention improved mitochondrial function. These core mecha-
and exercise appear to share many common mechanisms nisms appear to be central to improving DPN and contin-
relevant to DPN, including beneficial effects on obesity, ued study of these risk factors should help identify new
reductions in insulin resistance, reductions in glycemic pharmacological targets that can be targeted and coupled
levels, reductions in AGE accumulation, increased neuro- with existing medical treatments

3. Ang L, Jaiswal M, Martin C, Pop-Busui


targets. Exploration of these mechanisms merits
R.  Glucose control and diabetic neuropathy: les-
further research, particularly related to the over- sons from recent large clinical trials. Curr Diab
lapping mechanism between exercise and dietary Rep. 2014;14:528.
changes (Fig. 4). 4. Skyler JS.  Diabetic complications. The importance
of glucose control. Endocrinol Metab Clin N Am.
1996;25:243–54.
Acknowledgments This work was supported by NIH 5. Feldman EL, Nave KA, Jensen TS, Bennett
grants RO1 NS043314 (D.E.W.), T32 HD057850 (J.E. DLH.  New horizons in diabetic neuropathy:
and D.E.W.), the Kansas Institutional Development Award mechanisms, bioenergetics, and pain. Neuron.
(IDeA) P20 GM103418 (D.E.W.). All figures were cre- 2017;93:1296–313.
ated using BioRender (biorender.com). 6. American Diabetes A. 9. Pharmacologic
approaches to glycemic treatment: standards of
Conflict of Interest  The authors declare no medical care in diabetes-2020. Diabetes Care.
2020;43:S98–S110.
competing financial interests.
7. Mathieu C, Gillard P, Benhalima K.  Insulin ana-
logues in type 1 diabetes mellitus: getting better all
Author Contributions  J.E. and D.E.W. co- the time. Nat Rev Endocrinol. 2017;13:385–99.
wrote the chapter. 8. Tesfaye S, Boulton AJ, Dickenson AH. Mechanisms
and management of diabetic painful distal
symmetrical polyneuropathy. Diabetes Care.
2013;36:2456–65.
References 9. Oyibo SO, Prasad YD, Jackson NJ, Jude EB, Boulton
AJ. The relationship between blood glucose excur-
1. Coppini DV, Spruce MC, Thomas P, Masding sions and painful diabetic peripheral neuropathy: a
MG.  Established diabetic neuropathy seems irre- pilot study. Diabet Med. 2002;19:870–3.
versible despite improvements in metabolic and 10. Smith AG, Russell J, Feldman EL, Goldstein J, Peltier
vascular risk markers—a retrospective case-control A, Smith S, Hamwi J, Pollari D, Bixby B, Howard J,
study in a hospital patient cohort. Diabet Med. Singleton JR.  Lifestyle intervention for pre-diabetic
2006;23:1016–20. neuropathy. Diabetes Care. 2006;29:1294–9.
2. Zochodne DW.  The challenges of diabetic poly- 11. Callaghan BC, Xia R, Reynolds E, Banerjee M,
neuropathy: a brief update. Curr Opin Neurol. Rothberg AE, Burant CF, Villegas-Umana E,
2019;32:666–75. Pop-Busui R, Feldman EL.  Association between
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 409

metabolic syndrome components and polyneu- tance in diabetic neuropathy. Exp Diabetes Res.
ropathy in an obese population. JAMA Neurol. 2011;2011:212571.
2016;73:1468–76. 26. Guilford BL, Ryals JM, Lezi E, Swerdlow RH,
12. Feldman EL, Callaghan BC, Pop-Busui R, Zochodne Wright DE. Dorsal root ganglia mitochondrial bio-
DW, Wright DE, Bennett DL, Bril V, Russell JW, chemical changes in non-diabetic and streptozotocin-­
Viswanathan V.  Diabetic neuropathy. Nat Rev Dis induced diabetic mice fed with a standard or high-fat
Primers. 2019;5:41. diet. J Neurol Neurosci. 2017;8:180.
13. Agathos E, Tentolouris A, Eleftheriadou I, Katsaouni 27. Groover AL, Ryals JM, Guilford BL, Wilson NM,
P, Nemtzas I, Petrou A, Papanikolaou C, Tentolouris Christianson JA, Wright DE.  Exercise-mediated
N. Effect of alpha-lipoic acid on symptoms and qual- improvements in painful neuropathy associated with
ity of life in patients with painful diabetic neuropa- prediabetes in mice. Pain. 2013;154:2658–67.
thy. J Int Med Res. 2018;46:1779–90. 28. Coppey LJ, Shevalye H, Obrosov A, Davidson EP,
14. Papanas N, Ziegler D. Efficacy of alpha-lipoic acid Yorek MA. Determination of peripheral neuropathy
in diabetic neuropathy. Expert Opin Pharmacother. in high-fat diet fed low-dose streptozotocin-treated
2014;15:2721–31. female C57Bl/6J mice and Sprague-Dawley rats. J
15. Yang XD, Fang PF, Xiang DX, Yang YY.  Topical Diabetes Investig. 2018;9:1033–40.
treatments for diabetic neuropathic pain. Exp Ther 29. Cooper MA, Jack MM, Ryals JM, Hayley P, Escher
Med. 2019;17:1963–76. T, Koch LG, Britton SL, Raupp SM, Winter MK,
16. Heydari M, Homayouni K, Hashempur MH, Shams McCarson KE, Geiger PC, Thyfault JP, Wright
M.  Topical Citrullus colocynthis (bitter apple) DE.  Rats bred for low and high running capacity
extract oil in painful diabetic neuropathy: a double-­ display alterations in peripheral tissues and nerves
blind randomized placebo-controlled clinical trial. J relevant to neuropathy and pain. Brain Behav.
Diabetes. 2016;8:246–52. 2017;7:e00780.
17. Ostovar M, Akbari A, Anbardar MH, Iraji A, 30. Stavniichuk R, Drel VR, Shevalye H, Vareniuk
Salmanpour M, Hafez Ghoran S, Heydari M, I, Stevens MJ, Nadler JL, Obrosova IG.  Role of
Shams M.  Effects of Citrullus colocynthis L. in 12/15-lipoxygenase in nitrosative stress and periph-
a rat model of diabetic neuropathy. J Integr Med. eral prediabetic and diabetic neuropathies. Free
2020;18:59–67. Radic Biol Med. 2010;49:1036–45.
18. The Look AHEAD Research Group. Effects of a 31. Hubinont C, Sener A, Malaisse WJ. Sorbitol content
long-term lifestyle modification programme on of plasma and erythrocytes during induced short-­
peripheral neuropathy in overweight or obese adults term hyperglycemia. Clin Biochem. 1981;14:19–20.
with type 2 diabetes: the Look AHEAD study. 32. Stavniichuk R, Shevalye H, Hirooka H, Nadler
Diabetologia. 2017;60:980–8. JL, Obrosova IG.  Interplay of sorbitol pathway
19. Preguiça I, Alves A, Nunes S, Gomes P, Fernandes of glucose metabolism, 12/15-lipoxygenase, and
R, Viana SD, Reis F.  Diet-induced rodent models ­mitogen-­activated protein kinases in the pathogen-
of diabetic peripheral neuropathy, retinopathy and esis of diabetic peripheral neuropathy. Biochem
nephropathy. Nutrients. 2020;12:250. Pharmacol. 2012;83:932–40.
20. Guilford BL, Ryals JM, Wright DE.  Phenotypic 33. Bierhaus A, Fleming T, Stoyanov S, Leffler A, Babes
changes in diabetic neuropathy induced by a high-­ A, Neacsu C, Sauer SK, Eberhardt M, Schnölzer M,
fat diet in diabetic C57BL/6 mice. Exp Diabetes Res. Lasitschka F.  Methylglyoxal modification of Na v
2011;2011:848307. 1.8 facilitates nociceptive neuron firing and causes
21. Obrosova IG, Ilnytska O, Lyzogubov VV, Pavlov hyperalgesia in diabetic neuropathy. Nat Med.
IA, Mashtalir N, Nadler JL, Drel VR. High-fat diet 2012;18:926–33.
induced neuropathy of pre-diabetes and obesity: 34. Amano S, Yamagishi S-i, Kato N, Inagaki Y,
effects of "healthy" diet and aldose reductase inhibi- Okamoto T, Makino M, Taniko K, Hirooka H,
tion. Diabetes. 2007;56:2598–608. Jomori T, Takeuchi M. Sorbitol dehydrogenase over-
22. Stino AM, Smith AG. Peripheral neuropathy in pre- expression potentiates glucose toxicity to cultured
diabetes and the metabolic syndrome. J Diabetes retinal pericytes. Biochem Biophys Res Commun.
Investig. 2017;8:646–55. 2002;299:183–8.
23. Vincent AM, Hayes JM, McLean LL, Vivekanandan-­ 35. Kikuchi S, Shinpo K, Moriwaka F, Makita Z, Miyata
Giri A, Pennathur S, Feldman EL.  Dyslipidemia-­ T, Tashiro K.  Neurotoxicity of methylglyoxal and
induced neuropathy in mice: the role of oxLDL/ 3-deoxyglucosone on cultured cortical neurons:
LOX-1. Diabetes. 2009;58:2376–85. synergism between glycation and oxidative stress,
24. Grote CW, Groover AL, Ryals JM, Geiger PC, possibly involved in neurodegenerative diseases. J
Feldman EL, Wright DE. Peripheral nervous system Neurosci Res. 1999;57:280–9.
insulin resistance in ob/ob mice. Acta Neuropathol 36. Kuntz S, Kunz C, Rudloff S. Carbonyl compounds
Commun. 2013;1:15. methylglyoxal and glyoxal affect interleukin-8
25. Grote CW, Morris JK, Ryals JM, Geiger PC, secretion in intestinal cells by superoxide anion gen-
Wright DE.  Insulin receptor substrate 2 expres- eration and activation of MAPK p38. Mol Nutr Food
sion and involvement in neuronal insulin resis- Res. 2010;54:1458–67.
410 J. Enders and D. E. Wright

37. Polykretis P, Luchinat E, Boscaro F, Banci diagnosed type 2 diabetes: follow-up of a random-
L.  Methylglyoxal interaction with superoxide dis- ized trial. Diabetes Care. 2014;37:1824–30.
mutase 1. Redox Biol. 2020;30:101421. 50. Barrière DA, Noll C, Roussy G, Lizotte F, Kessai
38. Lupachyk S, Watcho P, Hasanova N, Julius U, A, Kirby K, Belleville K, Beaudet N, Longpré J-M,
Obrosova IG.  Triglyceride, nonesterified fatty Carpentier AC, Geraldes P, Sarret P.  Combination
acids, and prediabetic neuropathy: role for oxi- of high-fat/high-fructose diet and low-dose strepto-
dative–nitrosative stress. Free Radic Biol Med. zotocin to model long-term type-2 diabetes compli-
2012;52:1255–63. cations. Sci Rep. 2018;8:424.
39. Rumora AE, Lentz SI, Hinder LM, Jackson 51. García G, Gutiérrez-Lara EJ, Centurión D,
SW, Valesano A, Levinson GE, Feldman Granados-Soto V, Murbartián J.  Fructose-induced
EL.  Dyslipidemia impairs mitochondrial traffick- insulin resistance as a model of neuropathic pain in
ing and function in sensory neurons. FASEB J. rats. Neuroscience. 2019;404:233–45.
2018;32:195–207. 52. Rumora AE, LoGrasso G, Hayes JM, Mendelson FE,
40. Rumora AE, LoGrasso G, Haidar JA, Dolkowski Tabbey MA, Haidar JA, Lentz SI, Feldman EL. The
JJ, Lentz SI, Feldman EL.  Chain length of satu- divergent roles of dietary saturated and monounsatu-
rated fatty acids regulates mitochondrial traffick- rated fatty acids on nerve function in murine models
ing and function in sensory neurons. J Lipid Res. of obesity. J Neurosci. 2019;39:3770–81.
2019;60:58–70. 53. Tao M, McDowell MA, Saydah SH, Eberhardt
41. Viader A, Sasaki Y, Kim S, Strickland A, Workman MS. Relationship of polyunsaturated fatty acid intake
Cayce S, Yang K, Gross Richard W, Milbrandt to peripheral neuropathy among adults with diabetes
J.  Aberrant Schwann cell lipid metabolism linked in the National Health and Nutrition Examination
to mitochondrial deficits leads to axon degeneration Survey (NHANES) 1999–2004. Diabetes Care.
and neuropathy. Neuron. 2013;77:886–98. 2008;31:93.
42. Miyake T, Nakamura S, Zhao M, So K, Inoue K, 54. Brownlow ML, Benner L, D’Agostino D, Gordon
Numata T, Takahashi N, Shirakawa H, Mori Y, MN, Morgan D.  Ketogenic diet improves motor
Nakagawa T, Kaneko S. Cold sensitivity of TRPA1 performance but not cognition in two mouse
is unveiled by the prolyl hydroxylation blockade-­ models of Alzheimer’s pathology. PLoS One.
induced sensitization to ROS.  Nat Commun. 2013;8:e75713.
2016;7:12840. 55. Taylor MK, Sullivan DK, Mahnken JD, Burns JM,
43. Nazıroğlu M.  Activation of TRPM2 and TRPV1 Swerdlow RH. Feasibility and efficacy data from a
channels in dorsal root ganglion by NADPH oxidase ketogenic diet intervention in Alzheimer’s disease.
and protein kinase C molecular pathways: a patch Alzheimers Dement. 2018;4:28–36.
clamp study. J Mol Neurosci. 2017;61:425–35. 56. Bough KJ, Schwartzkroin PA, Rho JM.  Calorie
44. Nishio N, Taniguchi W, Sugimura YK, Takiguchi restriction and ketogenic diet diminish neuronal
N, Yamanaka M, Kiyoyuki Y, Yamada H, Miyazaki excitability in rat dentate gyrus in  vivo. Epilepsia.
N, Yoshida M, Nakatsuka T.  Reactive oxygen spe- 2003;44:752–60.
cies enhance excitatory synaptic transmission in rat 57. Dressler A, Trimmel-Schwahofer P, Klbermaxz K,
spinal dorsal horn neurons by activating TRPA1 and Prayer D, Kaspiran G, Rami B, Schober E, Feucht
TRPV1 channels. Neuroscience. 2013;247:201–12. M. Type 1 diabetes and epilepsy: efficacy and safety
45. Sözbir E, Nazıroğlu M. Diabetes enhances oxidative of the ketogenic diet. Epilepsia. 2010;51:1806–9.
stress-induced TRPM2 channel activity and its con- 58. Cooper MA, Menta BW, Perez-Sanchez C, Jack
trol by N-acetylcysteine in rat dorsal root ganglion MM, Khan ZW, Ryals JM, Winter M, Wright DE. A
and brain. Metab Brain Dis. 2016;31:385–93. ketogenic diet reduces metabolic syndrome-induced
46. Schink M, Leipold E, Schirmeyer J, Schönherr R, allodynia and promotes peripheral nerve growth in
Hoshi T, Heinemann SH.  Reactive species modify mice. Exp Neurol. 2018;306:149–57.
Na V 1.8 channels and affect action potentials in 59. Di Lorenzo C, Coppola G, Bracaglia M, Di Lenola
murine dorsal root ganglion neurons. Pflügers Arch. D, Sirianni G, Rossi P, Di Lorenzo G, Parisi V,
2016;468:99–110. Serrao M, Cervenka MC. A ketogenic diet normal-
47. Schlüter F, Leffler A.  Oxidation differentially izes interictal cortical but not subcortical responsiv-
modulates the recombinant voltage-gated Na+ ity in migraineurs. BMC Neurol. 2019;19:1–9.
channel α-subunits Nav1.7 and Nav1.8. Brain Res. 60. Ruskin DN, Sturdevant IC, Wyss LS, Masino
2016;1648:127–35. SA. Ketogenic diet effects on inflammatory allodynia
48. Esposito K, Maiorino M, Di Palo C, Giugliano D, and ongoing pain in rodents. Sci Rep. 2021;11:1–8.
Group CPHS.  Adherence to a Mediterranean diet 61. Holmden HF. Thesis on diabetes mellitus; its treat-
and glycaemic control in type 2 diabetes mellitus. ment by abstinence from food. Annexe thesis digiti-
Diabet Med. 2009;26:900–7. sation project 2016 Block 8. 1913.
49. Esposito K, Maiorino MI, Petrizzo M, Bellastella G, 62. Mosenthal HO, Clausen SW, Hiller A. The effect of
Giugliano D. The effects of a Mediterranean diet on diet on blood sugar in diabetes mellitus. Arch Intern
the need for diabetes drugs and remission of newly Med. 1918;XXI:93–108.
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 411

63. Newburgh L, Marsh PL. The use of a high fat diet in some–mediated inflammatory disease. Nat Med.
the treatment of diabetes mellitus: first paper. Arch 2015;21:263–9.
Intern Med. 1920;26:647–62. 77. Gris D, Ye Z, Iocca HA, Wen H, Craven RR,
64. Luukkonen PK, Dufour S, Lyu K, Zhang X-M, Gris P, Huang M, Schneider M, Miller SD, Ting
Hakkarainen A, Lehtimäki TE, Cline GW, Petersen JP-Y. NLRP3 plays a critical role in the development
KF, Shulman GI, Yki-Järvinen H. Effect of a keto- of experimental autoimmune encephalomyelitis by
genic diet on hepatic steatosis and hepatic mitochon- mediating Th1 and Th17 responses. J Immunol.
drial metabolism in nonalcoholic fatty liver disease. 2010;185:974–81.
Proc Natl Acad Sci. 2020;117:7347–54. 78. Hatscher L, Lehmann CHK, Purbojo A, Onderka
65. Chandrasekaran P, Rani PK.  Reversal of diabetic C, Liang C, Hartmann A, Cesnjevar R, Bruns H,
tractional retinal detachment attributed to keto diet. Gross O, Nimmerjahn F, Ivanović-Burmazović I,
BMJ Case Rep. 2020;13:e235873. Kunz M, Heger L, Dudziak D. Select hyperactivat-
66. Tóth CC, Z.  Type 1 diabetes mellitus successfully ing NLRP3 ligands enhance the T<sub>H</sub>1-
managed with the paleolithic ketogenic diet. Int J and T<sub>H</sub>17-inducing potential of human
Case Rep Images. 2014;5:699–703. type 2 conventional dendritic cells. Sci Signal.
67. Enders J, Swanson MT, Ryals J, Wright D.  A 2021;14:eabe1757.
ketogenic diet reduces mechanical allodynia and 79. Jia M, Wu C, Gao F, Xiang H, Sun N, Peng P, Li
improves epidermal innervation in diabetic mice. J, Yuan X, Li H, Meng X.  Activation of NLRP3
Pain. 2022;163(4):682–9. inflammasome in peripheral nerve contributes to
68. Hussain TA, Mathew TC, Dashti AA, Asfar S, paclitaxel-induced neuropathic pain. Mol Pain.
Al-Zaid N, Dashti HM. Effect of low-calorie versus 2017;13:1744806917719804.
low-carbohydrate ketogenic diet in type 2 diabetes. 80. Sweitzer S, Colburn R, Rutkowski M, DeLeo
Nutrition. 2012;28:1016–21. J.  Acute peripheral inflammation induces moder-
69. Poplawski MM, Mastaitis JW, Isoda F, Grosjean ate glial activation and spinal IL-1β expression that
F, Zheng F, Mobbs CV.  Reversal of diabetic correlates with pain behavior in the rat. Brain Res.
nephropathy by a ketogenic diet. PLoS One. 1999;829:209–21.
2011;6:e18604. 81. Sleiman SF, Henry J, Al-Haddad R, El Hayek L,
70. Bombeiro AL, Pereira BTN, Bonfanti AP, de Abou Haidar E, Stringer T, Ulja D, Karuppagounder
Oliveira ALR.  Immunomodulation by dimethyl SS, Holson EB, Ratan RR, Ninan I, Chao
fumarate treatment improves mouse sciatic nerve MV.  Exercise promotes the expression of brain
regeneration. Brain Res Bull. 2020;160:24–32. derived neurotrophic factor (BDNF) through the
71. Bonomo RR, Cook TM, Gavini CK, White CR, action of the ketone body β-hydroxybutyrate. eLife.
Jones JR, Bovo E, Zima AV, Brown IA, Dugas 2016;5:e15092.
LR, Zakharian E, Aubert G, Alonzo F, Calcutt NA, 82. Wang X, Wu X, Liu Q, Kong G, Zhou J, Jiang J, Wu
Mansuy-Aubert V.  Fecal transplantation and butyr- X, Huang Z, Su W, Zhu Q.  Ketogenic Metabolism
ate improve neuropathic pain, modify immune cell Inhibits Histone Deacetylase (HDAC) and reduces
profile, and gene expression in the PNS of obese oxidative stress after spinal cord injury in rats.
mice. Proc Natl Acad Sci. 2020;117:26482–93. Neuroscience. 2017;366:36–43.
72. Kong G, Huang Z, Ji W, Wang X, Liu J, Wu X, 83. Mikami D, Kobayashi M, Uwada J, Yazawa
Huang Z, Li R, Zhu Q.  The ketone metabolite T, Kamiyama K, Nishimori K, Nishikawa Y,
β-hydroxybutyrate attenuates oxidative stress in Nishikawa S, Yokoi S, Taniguchi T, Iwano M.
spinal cord injury by suppression of class I histone β-Hydroxybutyrate enhances the cytotoxic effect of
deacetylases. J Neurotrauma. 2017;34:2645–55. cisplatin via the inhibition of HDAC/survivin axis in
73. Shang S, Wang L, Zhang Y, Lu H, Lu X. The beta-­ human hepatocellular carcinoma cells. J Pharmacol
hydroxybutyrate suppresses the migration of glioma Sci. 2020;142:1–8.
cells by inhibition of NLRP3 inflammasome. Cell 84. Zou X, Meng J, Li L, Han W, Li C, Zhong R, Miao
Mol Neurobiol. 2018;38:1479–89. X, Cai J, Zhang Y, Zhu D. Acetoacetate accelerates
74. Puchalska P, Crawford PA. Multi-dimensional roles muscle regeneration and ameliorates muscular dys-
of ketone bodies in fuel metabolism, signaling, and trophy in mice*. J Biol Chem. 2016;291:2181–95.
therapeutics. Cell Metab. 2017;25:262–84. 85. Volek JS, Fernandez ML, Feinman RD, Phinney
75. Boccella S, Guida F, De Logu F, De Gregorio D, SD.  Dietary carbohydrate restriction induces a
Mazzitelli M, Belardo C, Iannotta M, Serra N, unique metabolic state positively affecting athero-
Nassini R, de Novellis V.  Ketones and pain: unex- genic dyslipidemia, fatty acid partitioning, and met-
plored role of hydroxyl carboxylic acid receptor abolic syndrome. Prog Lipid Res. 2008;47:307–18.
type 2  in the pathophysiology of neuropathic pain. 86. Goldhaber-Fiebert JD, Goldhaber-Fiebert SN,
FASEB J. 2019;33:1062–73. Tristán ML, Nathan DM.  Randomized controlled
76. Youm Y-H, Nguyen KY, Grant RW, Goldberg EL, community-based nutrition and exercise interven-
Bodogai M, Kim D, D'agostino D, Planavsky N, tion improves glycemia and cardiovascular risk fac-
Lupfer C, Kanneganti TD.  The ketone metabo- tors in type 2 diabetic patients in rural Costa Rica.
lite β-hydroxybutyrate blocks NLRP3 inflamma- Diabetes Care. 2003;26:24.
412 J. Enders and D. E. Wright

87. Luo J, Bavencoffe A, Yang P, Feng J, Yin S, Qian A, 101. Zhu L, Hao J, Cheng M, Zhang C, Huo C, Liu Y, Du
Yu W, Liu S, Gong X, Cai T. Zinc inhibits TRPV1 to W, Zhang X.  Hyperglycemia-induced Bcl-2/Bax-­
alleviate chemotherapy-induced neuropathic pain. J mediated apoptosis of Schwann cells via mTORC1/
Neurosci. 2018;38:474–83. S6K1 inhibition in diabetic peripheral neuropathy.
88. Ma W, Berg J, Yellen G. Ketogenic diet metabolites Exp Cell Res. 2018;367:186–95.
reduce firing in central neurons by opening KATP 102. Hao W, Tashiro S, Hasegawa T, Sato Y, Kobayashi
channels. J Neurosci. 2007;27:3618–25. T, Tando T, Katsuyama E, Fujie A, Watanabe R,
89. Marques EA, Mota J, Carvalho J.  Exercise effects Morita M, Miyamoto K, Morioka H, Nakamura M,
on bone mineral density in older adults: a meta-­ Matsumoto M, Amizuka N, Toyama Y, Miyamoto
analysis of randomized controlled trials. Age. T.  Hyperglycemia promotes schwann cell de-­
2012;34:1493–515. differentiation and de-myelination via sorbitol accu-
90. Andersson DA, Gentry C, Light E, Vastani N, mulation and Igf1 protein down-regulation*. J Biol
Vallortigara J, Bierhaus A, Fleming T, Bevan Chem. 2015;290:17106–15.
S.  Methylglyoxal evokes pain by stimulating 103. Tosaki T, Kamiya H, Yasuda Y, Naruse K, Kato K,
TRPA1. PLoS One. 2013;8:e77986. Kozakae M, Nakamura N, Shibata T, Hamada Y,
91. Kahya MC, Nazıroğlu M, Övey İS.  Modulation of Nakashima E, Oiso Y, Nakamura J.  Reduced NGF
diabetes-induced oxidative stress, apoptosis, and secretion by Schwann cells under the high glucose
Ca(2+) entry through TRPM2 and TRPV1 channels condition decreases neurite outgrowth of DRG neu-
in dorsal root ganglion and hippocampus of diabetic rons. Exp Neurol. 2008;213:381–7.
rats by melatonin and selenium. Mol Neurobiol. 104. Kawamura M, Ruskin DN, Masino SA.  Metabolic
2017;54:2345–60. autocrine regulation of neurons involves coop-
92. Griggs RB, Laird DE, Donahue RR, Fu W, Taylor eration among pannexin hemichannels, adenos-
BK.  Methylglyoxal requires AC1 and TRPA1 to ine receptors, and KATP channels. J Neurosci.
produce pain and spinal neuron activation. Front 2010;30:3886–95.
Neurosci. 2017;11:679. 105. Campanella ME, Chu H, Low PS.  Assembly and
93. Greco T, Glenn TC, Hovda DA, Prins ML. Ketogenic regulation of a glycolytic enzyme complex on the
diet decreases oxidative stress and improves mito- human erythrocyte membrane. Proc Natl Acad Sci.
chondrial respiratory complex activity. J Cereb 2005;102:2402–7.
Blood Flow Metab. 2016;36:1603–13. 106. Epstein T, Xu L, Gillies RJ, Gatenby RA. Separation
94. Cooper MA, McCoin C, Pei D, Thyfault JP, of metabolic supply and demand: aerobic glycoly-
Koestler D, Wright DE.  Reduced mitochondrial sis as a normal physiological response to fluctuating
reactive oxygen species production in peripheral energetic demands in the membrane. Cancer Metab.
nerves of mice fed a ketogenic diet. Exp Physiol. 2014;2:7.
2018;103:1206–12. 107. Puchalska P, Martin SE, Huang X, Lengfeld JE,
95. Cheng HT, Dauch JR, Porzio MT, Yanik BM, Hsieh Daniel B, Graham MJ, Han X, Nagy L, Patti GJ,
W, Smith AG, Singleton JR, Feldman EL. Increased Crawford PA. Hepatocyte-macrophage acetoacetate
axonal regeneration and swellings in intraepidermal shuttle protects against tissue fibrosis. Cell Metab.
nerve fibers characterize painful phenotypes of dia- 2019;29:383–398.e7.
betic neuropathy. J Pain. 2013;14:941–7. 108. Valdebenito R, Ruminot I, Garrido-Gerter P,
96. Christianson JA, Ryals JM, Johnson MS, Dobrowsky Fernández-Moncada I, Forero-Quintero L,
RT, Wright DE.  Neurotrophic modulation of Alegría K, Becker HM, Deitmer JW, Barros
myelinated cutaneous innervation and mechani- LF.  Targeting of astrocytic glucose metabolism by
cal sensory loss in diabetic mice. Neuroscience. beta-­hydroxybutyrate. J Cereb Blood Flow Metab.
2007;145:303–13. 2015;36:1813–22.
97. Hsieh Y-L, Lin C-L, Chiang H, Fu Y-S, Lue J-H, 109. Kawano T, Zoga V, McCallum JB, Wu HE, Gemes
Hsieh S-T.  Role of peptidergic nerve terminals in G, Liang MY, Abram S, Kwok WM, Hogan QH,
the skin: reversal of thermal sensation by calcitonin Sarantopoulos CD.  ATP-sensitive potassium cur-
gene-related peptide in TRPV1-depleted neuropa- rents in rat primary afferent neurons: biophysical,
thy. PLoS One. 2012;7:e50805. pharmacological properties, and alterations by pain-
98. Johnson MS, Ryals JM, Wright DE.  Early loss of ful nerve injury. Neuroscience. 2009;162:431–43.
peptidergic intraepidermal nerve fibers in an STZ-­ 110. Sarantopoulos C, McCallum B, Sapunar D, Kwok
induced mouse model of insensate diabetic neuropa- W-M, Hogan Q.  ATP-sensitive potassium chan-
thy. Pain. 2008;140:35–47. nels in rat primary afferent neurons: the effect of
99. Johnson PC, Doll SC, Cromey DW. Pathogenesis of neuropathic injury and gabapentin. Neurosci Lett.
diabetic neuropathy. Ann Neurol. 1986;19:450–7. 2003;343:185–9.
100. Malik RA, Tesfaye S, Newrick PG, Walker D, 111. Wu X-F, Liu W-T, Liu Y-P, Huang Z-J, Zhang Y-K,
Rajbhandari SM, Siddique I, Sharma AK, Boulton Song X-J.  Reopening of ATP-sensitive potassium
AJM, King RHM, Thomas PK, Ward JD. Sural nerve channels reduces neuropathic pain and regulates
pathology in diabetic patients with minimal but pro- astroglial gap junctions in the rat spinal cord. Pain.
gressive neuropathy. Diabetologia. 2005;48:578–85. 2011;152:2605–15.
Lifestyle and Dietary Modifications: Relevance in the Management of Diabetic Neuropathy 413

112. Xia H, Zhang D, Yang S, Wang Y, Xu L, Wu J, Ren ment of nerve-injury induced mechanical hyper-
J, Yao W, Fan L, Zhang C, Tian Y, Pan H-L, Wang sensitivity in female mice. Brain Behav Immun.
X.  Role of ATP-sensitive potassium channels in 2021;97:42–60.
modulating nociception in rat model of bone cancer 124. Chen T, Li H, Yin Y, Zhang Y, Liu Z, Liu
pain. Brain Res. 2014;1554:29–35. H. Interactions of Notch1 and TLR4 signaling path-
113. Fernandes VHR, Chaves FRP, Soares AAS, Breder ways in DRG neurons of in vivo and in vitro models
I, Kimura-Medorima ST, Munhoz DB, Cintra RMR, of diabetic neuropathy. Sci Rep. 2017;7:1–12.
Breder JC, Barreto J, Nadruz W, Carvalho LSF, 125. Reichardt F, Chassaing B, Nezami BG, Li G,
Quinaglia T, Arieta CEL, Sposito AC. Dapagliflozin Tabatabavakili S, Mwangi S, Uppal K, Liang B,
increases retinal thickness in type 2 diabetic patients Vijay-Kumar M, Jones D.  Western diet induces
as compared with glibenclamide: a randomized con- colonic nitrergic myenteric neuropathy and dys-
trolled trial. Diabetes Metab. 2021;47:101280. motility in mice via saturated fatty acid-and
114. Henquin JC.  ATP-sensitive K+ channels may con- lipopolysaccharide-­ induced TLR4 signalling. J
trol glucose-induced electrical activity in pan- Physiol. 2017;595:1831–46.
creatic B-cells. Biochem Biophys Res Commun. 126. Li Y, Zhang H, Kosturakis AK, Cassidy RM, Zhang
1988;156:769–75. H, Kennamer-Chapman RM, Jawad AB, Colomand
115. Henquin JC. Role of voltage- and Ca2(+)-dependent CM, Harrison DS, Dougherty PM.  MAPK signal-
K+ channels in the control of glucose-induced elec- ing downstream to TLR4 contributes to paclitaxel-­
trical activity in pancreatic B-cells. Pflugers Arch. induced peripheral neuropathy. Brain Behav Immun.
1990;416:568–72. 2015;49:255–66.
116. de Campos Lima T, DdO S, JBP L, Chiovato LM, 127. Louis P, Flint HJ.  Formation of propionate and
CMdC L.  Hyperglycemia induces mechanical butyrate by the human colonic microbiota. Environ
hyperalgesia and depolarization of the resting mem- Microbiol. 2017;19:29–41.
brane potential of primary nociceptive neurons: Role 128. Kim HJ, Leeds P, Chuang DM. The HDAC inhibi-
of ATP-sensitive potassium channels. J Neurol Sci. tor, sodium butyrate, stimulates neurogenesis in the
2019;401:55–61. ischemic brain. J Neurochem. 2009;110:1226–40.
117. Ruan J-S, Lin J-K, Kuo Y-Y, Chen Y-W, Chen 129. Tong X, Yin L, Giardina C.  Butyrate suppresses
P-C.  Chronic palmitic acid-induced lipotoxicity Cox-2 activation in colon cancer cells through
correlates with defective trafficking of ATP sensi- HDAC inhibition. Biochem Biophys Res Commun.
tive potassium channels in pancreatic β cells. J Nutr 2004;317:463–71.
Biochem. 2018;59:37–48. 130. Liu C, Li C, Deng Z, Du E, Xu C. Long non-coding
118. Feinman RD, Volek JS.  Low carbohydrate diets RNA BC168687 is involved in TRPV1-mediated
improve atherogenic dyslipidemia even in the diabetic neuropathic pain in rats. Neuroscience.
absence of weight loss. Nutr Metab. 2006;3:24. 2018;374:214–22.
119. Kesl SL, Poff AM, Ward NP, Fiorelli TN, Ari C, 131. Lindefeldt M, Eng A, Darban H, Bjerkner A,
Van Putten AJ, Sherwood JW, Arnold P, D’Agostino Zetterström CK, Allander T, Andersson B,
DP.  Effects of exogenous ketone supplementation Borenstein E, Dahlin M, Prast-Nielsen S. The keto-
on blood ketone, glucose, triglyceride, and lipo- genic diet influences taxonomic and functional com-
protein levels in Sprague–Dawley rats. Nutr Metab. position of the gut microbiota in children with severe
2016;13:9. epilepsy. NPJ Biofilms Microbiomes. 2019;5:5.
120. Kephart WC, Mumford PW, Mao X, Romero MA, 132. Ferraris C, Meroni E, Casiraghi MC, Tagliabue A,
Hyatt HW, Zhang Y, Mobley CB, Quindry JC, Young De Giorgis V, Erba D. One month of classic thera-
KC, Beck DT.  The 1-week and 8-month effects of peutic ketogenic diet decreases short chain fatty
a ketogenic diet or ketone salt supplementation on acids production in epileptic patients. Front Nutr.
multi-organ markers of oxidative stress and mito- 2021;8:613100.
chondrial function in rats. Nutrients. 2017;9:1019. 133. Nagpal R, Neth BJ, Wang S, Craft S, Yadav
121. Shen S, Lim G, You Z, Ding W, Huang P, Ran H.  Modified Mediterranean-ketogenic diet modu-
C, Doheny J, Caravan P, Tate S, Hu K, Kim H, lates gut microbiome and short-chain fatty acids
McCabe M, Huang B, Xie Z, Kwon D, Chen L, in association with Alzheimer's disease mark-
Mao J.  Gut microbiota is critical for the induc- ers in subjects with mild cognitive impairment.
tion of chemotherapy-­induced pain. Nat Neurosci. EBioMedicine. 2019;47:529–42.
2017;20:1213–6. 134. Cuff DJ, Meneilly GS, Martin A, Ignaszewski A,
122. Ramakrishna C, Corleto J, Ruegger PM, Logan GD, Tildesley HD, Frohlich JJ. Effective exercise modal-
Peacock BB, Mendonca S, Yamaki S, Adamson ity to reduce insulin resistance in women with type 2
T, Ermel R, McKemy D, Borneman J, Cantin diabetes. Diabetes Care. 2003;26:2977.
EM.  Dominant role of the gut microbiota in che- 135. Dall'aglio E, Chang F, Chang H, Wright D, Reaven
motherapy induced neuropathic pain. Sci Rep. GM. Effect of exercise training and sucrose feeding
2019;9:20324. on insulin-stimulated glucose uptake in rats with
123. Szabo-Pardi TA, Barron LR, Lenert ME, Burton streptozotocin-induced insulin-deficient diabetes.
MD.  Sensory neuron TLR4 mediates the develop- Diabetes. 1983;32:165–8.
414 J. Enders and D. E. Wright

136. Kluding PM, Pasnoor M, Singh R, Jernigan S, large-fiber neuropathy in diabetic mice. Diabetes.
Farmer K, Rucker J, Sharma NK, Wright DE.  The 2008;57:1693.
effect of exercise on neuropathic symptoms, nerve 147. van Deursen RWM, Sanchez MM, Ulbrecht JS,
function, and cutaneous innervation in people Cavanagh PR. The role of muscle spindles in ankle
with diabetic peripheral neuropathy. J Diabetes movement perception in human subjects with dia-
Complicat. 2012;26:424–9. betic neuropathy. Exp Brain Res. 1998;120:1–8.
137. Balducci S, Iacobellis G, Parisi L, Di Biase N, 148. Allet L, Armand S, de Bie RA, Golay A, Monnin
Calandriello E, Leonetti F, Fallucca F.  Exercise D, Aminian K, Staal JB, de Bruin ED. The gait and
training can modify the natural history of dia- balance of patients with diabetes can be improved:
betic peripheral neuropathy. J Diabetes Complicat. a randomised controlled trial. Diabetologia.
2006;20:216–23. 2010;53:458–66.
138. Oh TJ, Lee J-E, Choi SH, Jang HC.  Association 149. Singleton JR, Marcus RL, Jackson JE, Lessard M,
between body fat and diabetic peripheral neuropa- Graham TE, Smith AG.  Exercise increases cutane-
thy in middle-aged adults with type 2 diabetes mel- ous nerve density in diabetic patients without neu-
litus: a preliminary report. J Obes Metab Syndr. ropathy. Ann Clin Transl Neurol. 2014;1:844–9.
2019;28:112–7. 150. Roy Chowdhury SK, Smith DR, Saleh A,
139. Bassey EJ, Rothwell MC, Littlewood JJ, Pye Schapansky J, Marquez A, Gomes S, Akude E,
DW.  Pre- and postmenopausal women have dif- Morrow D, Calcutt NA, Fernyhough P.  Impaired
ferent bone mineral density responses to the adenosine monophosphate-activated protein kinase
same high-impact exercise. J Bone Miner Res. signalling in dorsal root ganglia neurons is linked to
1998;13:1805–13. mitochondrial dysfunction and peripheral neuropa-
140. Kelley GA, Kelley KS, Tran ZV. Exercise and bone thy in diabetes. Brain. 2012;135:1751–66.
mineral density in men: a meta-analysis. J Appl 151. Yoon H, Thakur V, Isham D, Fayad M, Chattopadhyay
Physiol. 2000;88:1730–6. M. Moderate exercise training attenuates inflamma-
141. Strotmeyer ES, Cauley JA, Schwartz AV, de tory mediators in DRG of Type 1 diabetic rats. Exp
Rekeneire N, Resnick HE, Zmuda JM, Shorr RI, Neurol. 2015;267:107–14.
Tylavsky FA, Vinik AI, Harris TB. Reduced periph- 152. Zheng Q, Huang C, Guo J, Tan J, Wang C, Tang B,
eral nerve function is related to lower hip BMD and Zhang H.  Hsp70 participates in PINK1-mediated
calcaneal QUS in older white and black adults: the mitophagy by regulating the stability of PINK1.
Health, Aging, and Body Composition Study. J Bone Neurosci Lett. 2018;662:264–70.
Miner Res. 2006;21:1803–10. 153. Schnyder S, Svensson K, Cardel B, Handschin
142. Lombardo YB, Hein G, Chicco A.  Metabolic syn- C. Muscle PGC-1α is required for long-term systemic
drome: effects of n-3 PUFAs on a model of dys- and local adaptations to a ketogenic diet in mice. Am
lipidemia, insulin resistance and adiposity. Lipids. J Physiol Endocrinol Metab. 2017;312:E437–46.
2007;42:427–37. 154. Wrann CD, White JP, Salogiannnis J, Laznik-­
143. Semple RK, Sleigh A, Murgatroyd PR, Adams CA, Bogoslavski D, Wu J, Ma D, Lin JD, Greenberg
Bluck L, Jackson S, Vottero A, Kanabar D, Charlton-­ ME, Spiegelman BM.  Exercise induces hippocam-
Menys V, Durrington P, Soos MA, Carpenter TA, pal BDNF through a PGC-1α/FNDC5 pathway. Cell
Lomas DJ, Cochran EK, Gorden P, O’Rahilly S, Metab. 2013;18:649–59.
Savage DB. Postreceptor insulin resistance contrib- 155. Chandrasekaran K, Anjaneyulu M, Inoue T, Choi J,
utes to human dyslipidemia and hepatic steatosis. J Sagi AR, Chen C, Ide T, Russell JW. Mitochondrial
Clin Invest. 2009;119:315–22. transcription factor A regulation of mitochondrial
144. Gómez-Pinilla F, Ying Z, Opazo P, Roy R, Edgerton degeneration in experimental diabetic neuropathy.
V. Differential regulation by exercise of BDNF and Am J Physiol Endocrinol Metab. 2015;309:E132–41.
NT-3  in rat spinal cord and skeletal muscle. Eur J 156. Chandrasekaran K, Salimian M, Konduru SR, Choi
Neurosci. 2001;13:1078–84. J, Kumar P, Long A, Klimova N, Ho C-Y, Kristian T,
145. Feczko JD, Klueber KM. Cytoarchitecture of muscle Russell JW.  Overexpression of Sirtuin 1 protein in
in a genetic model of murine diabetes. Am J Anat. neurons prevents and reverses experimental diabetic
1988;182:224–40. neuropathy. Brain. 2019;142:3737–52.
146. Muller KA, Ryals JM, Feldman EL, Wright
DE.  Abnormal muscle spindle innervation and
Pathophysiology of Neuropathic
Pain

Andreas C. Themistocleous
and Miroslav Misha Backonja

1 Introduction and pins-and-needles, i.e. paraesthesias or dys-


aesthesias. In diabetic neuropathy, the pattern of
Neuropathic pain is defined as “pain arising from neuropathic pain depends on the nerves affected.
disease or lesion of somatosensory nervous sys- In distal symmetric polyneuropathy, the most
tem” [1]. It is a heterogenous condition in which common form of diabetic neuropathy, neuro-
the aetiology of the inciting nerve lesion interacts pathic pain is found in the toes, feet, and legs that
with individual contributory factors such as gen- can progress to involve the fingers and hands [4],
otype and environmental factors [2]. The indi- thus evolving into a “glove-and-stocking” distri-
vidual neuropathic pain phenotype is the end bution synonymous with diabetic neuropathy.
result of combinations of pathophysiological A number of risk factors from large cross-­
mechanisms that include sensory nerve damage, sectional cohort studies are linked to painful dia-
ectopic activity, sensitisation of the peripheral betic neuropathy (Table  1). Poorer diabetic
and central nervous system, and altered brain control, obesity, polyneuropathy severity,
connectivity. A single aetiology, such as diabetes increasing age, female gender, and ethnicity are
mellitus, has a broad range of pathological pre- possible risk factors for development and persis-
sentations and does not cause neuropathic pain in tence of painful diabetic neuropathy [5–9]. The
a uniform way. correlation of these risk factors to neuropathic
Neuropathic pain is a common complication pain is not linear, as patients with severe neuropa-
caused by diabetic neuropathy. It affects between
25 and 50% of patients, depending on study
inclusion criteria [3]. Painful diabetic neuropathy Table 1  Risk factors for development of painful symp-
is often described as burning. Other descriptors toms in diabetic polyneuropathy
include stabbing, shooting, stinging, or aching Demographic factors
sensation and are often associated with tingling  • Advancing age
 • Female sex
 • Ethnicity
Clinical
A. C. Themistocleous  • Poorer diabetic control
Nuffield Department of Clinical Neurosciences,  • Polyneuropathy severity
University of Oxford, Oxford, UK  • Obesity
e-mail: andreas.themistocleous@ndcn.ox.ac.uk Genetics
 • Heritability of pain traits
M. M. Backonja (*)
 • Voltage gated sodium channels
NCCIH, NIH, Bethesda, MD, USA
e-mail: misha.backonja@nih.gov Psychological factors: Depression, anxiety

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 415
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_23
416 A. C. Themistocleous and M. M. Backonja

thy or very poor diabetic control can have pain- 2 Peripheral Mechanisms
less diabetic neuropathy. Several studies have of Painful Diabetic
shown associations between genotype and the Neuropathy
risk of developing neuropathic pain [10].
Although a number of findings still need to be In human studies, microneurography recordings
replicated, the heritability of pain traits shows in painful diabetic neuropathy have shown spon-
that genetics has an important role [11]. taneous activity in nociceptive fibres, mechano-­
Psychological factors are emerging as critical insensitive C fibre sensitisation, altered
factors in the expression of chronic painful dia- distribution of C afferent nerve fibres, and loss of
betic neuropathy. The processing of peripheral function within polymodal nociceptors (Table 2)
nociceptive signals by brain networks involved in [15–17]. However, in the only study comparing
processing pain related experiences will vary microneurography recordings between patients
between individuals and shape how individuals with painful and painless diabetic distal symmet-
experience, report, and manage chronic pain. rical polyneuropathy, some degree of spontane-
Longitudinal prospective natural history studies, ous activity and sensitisation was found in
combined with reliable measures of relevant patients with painless diabetic neuropathy [16].
pathophysiological mechanisms are required to This observation does not have clear interpreta-
explore and understand the relationship between tion, as it was a small study of only eight partici-
risk factors and pain in diabetic neuropathy. pants, of which five were diagnosed with
However, these studies are yet to be done. neuropathic pain. An important assumption based
A paradox of painful diabetic neuropathy is on other human neuropathic pain microneurogra-
the combination of ongoing, stimulus-­phy studies is that spontaneous activity and sensi-
independent neuropathic pain and sensory loss in tisation are important for the development and
a painful area. A typical clinical presentation is maintenance of neuropathic pain [18, 19]. C fibre
spontaneous ongoing pain and numbness with polymodal receptors with spontaneous activity
hyposensitivity to evoked pinprick, touch, and and altered stimulus-response function properties
temperature stimuli. Detailed studies that com- (lowered response threshold, increased response
bine interviews, clinical assessment, and quanti- magnitude, and a less adaptive response pattern
tative sensory testing show that sensory loss is to mechanical stimulation) are seen in rodent
the most common sensory phenotype in diabetic
polyneuropathy [6, 8, 9]. Mechanical hypersensi- Table 2 Neuronal and non-neuronal contributor to
tivity to evoked stimuli, i.e. mechanical hyperal- peripheral neuropathic pain mechanisms
gesia or allodynia, is an uncommon finding, Neuronal factors
present in only 15% of study participants [8].  • Spontaneous activity due to abnormal ectopic
However, dynamic mechanical allodynia is the activity
only discriminator between painless and painful  • Mechano-insensitive C fibres sensitisation
 • Altered distribution of C afferent nerve fibres
diabetic neuropathy, found only in those with  • Polymodal nociceptor loss function
neuropathic pain [12]. This contrasts to other  • Increased voltage gated sodium expression—
neuropathic pain conditions in which mechanical enhanced currents
hypersensitivity is a common finding and can  • Post-translational modification of ion channels, e.g.
methylglyoxal
guide treatment [13, 14].  • TRPA1 activation
In the following section we will discuss the pos-  • Peripheral neuronal sensitisation of TRPV1 in dorsal
sible pathophysiological mechanisms, within the root ganglion neurons
peripheral and central nervous systems, that cause  • Increased CaV3.2 T-type channel activity
Non-neuronal factors
painful diabetic neuropathy. The majority of mech-  • Higher epineural oxygen saturation
anisms discussed will apply to distal symmetrical  • Faster epineural blood flow—arterio-venous
polyneuropathy because it is the most common shunting
presentation of painful diabetic neuropathy.  • Pro-inflammatory cytokines
Pathophysiology of Neuropathic Pain 417

models of diabetic neuropathy [18, 20]. frequency firing of action potentials. NaV 1.9
Therefore, abnormal ectopic activity and periph- amplifies subthreshold depolarisation, the large
eral nociceptor sensitisation are pathophysiologi- window current may contribute to resting mem-
cal hallmarks of nerve injury and aberrant brane potential. NaV 1.3, when expressed after
signalling within nociceptive fibres. peripheral nerve injury, amplifies subthreshold
Abnormal ectopic activity can occur at multi- depolarisation, and rapid repriming supports
ple sites that include site of nerve injury, periph- high-frequency firing of action potentials. In
eral axon and dorsal root ganglia of nociceptive experimental rodent models of diabetic neuropa-
neurones [21]. Sensory neuronal excitability is thy, NaV1.8 expression is increased and enhanced
controlled by the expression, trafficking, and currents mediated by the channel reduce C fibre
function of ligand and voltage gated ion chan- nociceptor conduction failure, which leads to
nels. These channels are responsible for sensory increased neuronal conduction to the central ner-
stimuli transduction, action potential generation vous system [25].
and propagation, and neurotransmitter release Emerging evidence shows that inherited NaV
with the dorsal horn. Voltage gated sodium (NaV) 1.7 and NaV 1.8 mutations that cause dorsal root
channels are important drivers of excitability ganglion hyperexcitability may increase the risk
within nociceptor neurones, as they are crucial for the development of painful diabetic neuropa-
for action potential electrogenesis. Clustering of thy [26–29]. Gain of function variants may also
voltage gated sodium channels at sites of injury cause axonal degeneration through increased
lowers the action potential thresholds and causes energetic demand and reactive oxygen species
an increase in excitability within primary production [30, 31]. The G856D mutation in
afferents. NaV1.7 produces significant changes in channel
Nine different NaV channels (NaV1.1–NaV1.9) biophysical properties, which contribute to the
are expressed in humans and each has a unique hyperexcitability exhibited by neurones contain-
pattern of expression across different types of ing Nav1.8. The biophysical channel changes
excitable cells. The sodium channels NaV 1.7, lead to ATP depletion, increased reactive oxygen
1.8, and 1.9 are the most highly expressed species, elevated intracellular Na+ and Ca2+.
α-subunits in nociceptors [22]. NaV 1.3 is These changes cause increased axonal depolari-
expressed in embryonic tissue only. However, sation with subsequent axonal degeneration.
NaV 1.3 expression is significantly upregulated Blockade of the reverse mode of the sodium/cal-
after nerve injury [23]. Rare extreme inherited cium exchanger protects against neurite degen-
channelopathies show the importance of voltage eration. This variant has not been studied in the
gated sodium channel in nociceptive pathways context of diabetes, but it is conceivable that the
[24]. For example, loss of function mutations in energetic demands of diabetes may amplify the
NaV 1.7 causes congenital insensitivity to pain, changes caused by G856D mutation in NaV1.7.
while gain of function mutations causes erythro- The role of NaV 1.9 and NaV 1.3 has not been
melalgia and paroxysmal extreme pain disorder. studied in painful diabetic neuropathy and their
Patch clamp recordings from small diameter dor- role is unclear.
sal root ganglion neurones show that Na  +  cur- Post-translational modification of ion chan-
rent characteristics vary between neurones. These nels, caused by metabolites that increase in dia-
experiments show that subunit expression and betes, is an important potential mechanism in
post-translational modification are important in neuropathic pain. Methylglyoxal is a highly reac-
regulating neuronal firing. NaV 1.7 is a threshold tive metabolite that increases in diabetes and
channel for action potential generation, amplifies causes non-enzymatic glycation of proteins [32].
subthreshold depolarisation, and contributes to Furthermore, the enzyme that metabolises meth-
the rising phase of the action potential. NaV 1.8 is ylglyoxal, gyloxylase-1, is lowly expressed in the
the main contributor to the rising phase of action peripheral nervous system, and activity is further
potentials, and rapid repriming supports high-­ reduced in diabetes. In human microneurography
418 A. C. Themistocleous and M. M. Backonja

studies methylglyoxal causes pain, axon-reflex TRPV1 attenuate nociceptive behaviours and
erythema, and long lasting hyperalgesia through thermal hyperalgesia in animal models of neuro-
activation of mechano-insensitive C fibres [33]. pathic pain [42–44]. Changes in the expression of
In vitro data shows that methylglyoxal directly TRPV1, in rodent diabetic neuropathy models,
activates cultured dorsal root ganglia nocicep- correlate with mechanical allodynia [45], devel-
tors, evokes the release of calcitonin-gene related opment of thermal hyperalgesia and TRPV1
peptide, and causes pain hypersensitivity in expression on large myelinated A-fibres, an
rodents by post-translational modification of injury-induced phenotypic shifts in low-­threshold
NaV1.8 that reduces inactivation of the channel sensory neurones [46]. A possible mechanism of
with subsequent nociceptor hyperexcitability TRPV1 mediated neuropathic pain is peripheral
[34]. neuronal sensitisation of TRPV1  in dorsal root
Other channels implicated in painful diabetic ganglion neurones [47]. High serum glucose con-
neuropathy include: transient receptor potential centration and the inflammatory cytokine high-­
cation channel, subfamily A member 1 (TRPA1); mobility group box-protein 1, which is
nonselective transient receptor potential cation upregulated in the skin of diabetic rats, enhance
channel subfamily V member 1 (TRPV1); and TRPV1-evoked Ca2+ responses in dorsal gan-
T-type Ca2+ channels. glion neurones. This is achieved through the
TRPA1 is a nonselective ligand gated cation receptor for advanced glycation end products in a
channel expressed in a subset of nociceptors. It is protein kinase C dependent manner and is
activated by exogenous compounds such as mus- blocked by vascular endothelial growth factor
tard oil and cinnamaldehyde, noxious cold, and [47]. In human studies, pre-injection with a
endogenous ligands such as prostaglandins [35]. TRPV1 antagonist does not block pain sensation
TRPA1 is implicated in painful diabetic neuropa- and hyperalgesia after methylglyoxal administra-
thy. In rodent models of streptozotocin induced tion [33].
diabetic neuropathy, pain related behaviour was T-type Ca2+channels regulate the subthreshold
attenuated after administration of a TRPA1 chan- excitability of nociceptors [48].
nel antagonist [36]. However, a major caveat of In streptozotocin models of diabetic neuropa-
this model is that streptozotocin directly activates thy, CaV3.2 T-type channel activity is increased.
TRPA1 channels [37]. In human studies, pre-­ Glycosylation of extracellular arginine residues
injection with a TRPA1 antagonist blocks pain of CaV3.2  T-type channel causes increased cur-
sensation and hyperalgesia after methylglyoxal rent density, increased surface expression, and
administration [33]. Methylglyoxal binds and altered kinetics [49–51]. All of which contribute
modifies intracellular cysteine residues that acti- to dorsal root ganglion hyperexcitability.
vate TRPA1 with resultant hyperexcitability of Furthermore, downregulation of CaV3.2  T-type
nociceptive neurones [38]. The reactive oxygen channel improves pain related behaviours in
species hydrogen peroxide and 4-­hydroxyhexenal, streptozotocin model of rodent diabetic neuropa-
which are increased in diabetes, also activate thy [50, 51].
TRPA1 [35]. Although further validation is
required, TRPA1 is a promising therapeutic
target. 2.1 Endothelial Dysfunction
TRPV1 is a nonselective ligand gate cation and Inflammation
channel that is activated by endogenous and
exogenous chemicals such as capsaicin and high Nerve blood flow impairment, hypoxia, and car-
noxious temperatures. Its role in peripheral diovascular risk factors are important in the
inflammatory pain is well characterised [39]. pathogenesis of diabetic neuropathy [52]. Nerve
After partial nerve injury, TRPV1 is upregulated haemodynamics differ between patients with
in uninjured sensory fibres [40, 41], and inhibi- painless and painful diabetics neuropathy [53,
tion of TRPV1 activity or reducing levels of 54]. Higher epineural oxygen saturation and
Pathophysiology of Neuropathic Pain 419

faster epineural blood flow are present in those Table 3  Neuronal and non-neuronal contributor to cen-
with painful diabetic neuropathy [54]. These tral neuropathic pain mechanisms
findings may be related to arterio-venous shunt- Neuronal factors
 • Exaggerated response to synaptic input
ing, but it is not clear how haemodynamic  • Lower threshold for neuronal activation
changes contribute to neuropathic pain. Skin  • Increased response to suprathreshold stimuli
microvascular vasodilator and vasoconstrictor  • Receptive field expansion
responses are altered in diabetic neuropathy;  • Morphological alterations in postsynaptic structures
within second-order sensory neurons
however, no consistent relationship to painful  • Dysregulation of potassium-chloride cotransporter 2
diabetic neuropathy has been found. Pro-­ activity
inflammatory cytokines predict the incidence and  • Decreased pain thresholds and wide dynamic range
progression of distal sensorimotor polyneuropa- neuron hyperexcitability
thy [55], and serum markers of endothelial dys- Non-neuronal contributors to central neuropathic pain
mechanisms
function and inflammation are elevated in patients  • Segmental microglial activation
with painful diabetic neuropathy [56, 57]. In  • Neuroinflammation
rodent models of painful diabetic neuropathy,
pro-inflammatory cytokines expression is ele-
vated in serum and dorsal root ganglia neurones microglial activation, and neuroinflammation in
[58, 59]. These data suggest a link between local the central nervous system [61–63]. In strepto-
blood flood dysregulation, inflammation, and zotocin induced diabetic neuropathy dendritic
painful diabetic neuropathy. Further studies are spine remodelling was related to decreased pain
required to better understand these relationships thresholds and wide dynamic range neuron
and delineate the underlying mechanisms. hyperexcitability [60]. A Rac1-specific inhibitor
In summary, there is a clear role for ion chan- known to interfere with spine plasticity decreased
nel dysfunction in the pathogenesis of painful the presence of malformed spines, attenuated
diabetic neuropathy. The increase in neuronal neuronal hyperresponsiveness to peripheral stim-
activity from sensitised nociceptors and ectopic uli, reduced spontaneous firing activity from
foci in degenerating nerve fibres will cause wide dynamic range neurones, and improved
increased synaptic transmission within the dorsal nociceptive mechanical pain thresholds [60]. The
horn of the spinal cord. The phenomenon of cen- role of dorsal horn microglia has been studied
tral sensitisation will be discussed in the next through the administration of a minocycline, a
section. selective inhibitor of microglial activation, in
streptozotocin induced neuropathy in rats.
Administration of minocycline attenuated the
3 Central Mechanisms development of diabetic neuropathy and pain
of Painful Diabetic behaviours and reduced levels of interleukin-1β
Neuropathy and tumour necrosis factor-α, lipid peroxidation,
and nitrite within the spinal cord [62]. In a similar
Central sensitisation of dorsal horn neurones is experiment minocycline reduced the expression
characterised by exaggerated response to synap- of the potassium chloride cotransporter 2 within
tic input, lower threshold for neuronal activation, the spinal cord [61]. Inhibition of microglial acti-
increased response to suprathreshold stimuli, and vation through inhibition of extracellular signal-­
receptive field expansion (Table 3) [14]. There is regulated protein kinase signalling ameliorated
emerging evidence that similar mechanisms play allodynia is a streptozotocin induced diabetic
a role in painful diabetic neuropathy. Possible neuropathy [63]. Microglia release several cyto-
mechanisms include morphological alterations in kines and chemokines that increase dorsal horn
postsynaptic structures within second-order sen- neurone excitability after nerve injury. For exam-
sory neurons [60], dysregulation of potassium-­ ple, the ionotropic ATP receptor, P2X4, in
chloride cotransporter 2 activity [61], segmental microglial cells upregulates after nerve injury.
420 A. C. Themistocleous and M. M. Backonja

After upregulation, ATP-induced activation of grey mediated descending pain modulatory sys-
P2X4 on microglia results in the release of brain tem may signal a brain-based pain facilitation
derived neurotrophic factor (BDNF) from these mechanism for painful diabetic polyneuropathy.
cells. BDNF via its receptor, TrkB present on In streptozotocin rodent models’ electrophysio-
dorsal horn neurones, decreases K+/Cl− cotrans- logical analysis of the rostroventromedial
porter KCC2 activity [64]. The net effect being medulla shows an increase in the spontaneous
dorsal horn neurone excitability. The role of activity of pronociceptive ON-like cells and
NMDA receptors in painful diabetic neuropathy decreased spontaneous activity of OFF-like cells
is unclear. In animal models NMDA receptors [71]. These changes are similar to traumatic neu-
show increased expression and activation within ropathic pain models and suggest that enhanced
the spinal cord; however, NMDA receptor antag- descending pain facilitation may contribute to
onist drug trials in patients have shown limited rodent pain behaviours. Immunohistochemistry
efficacy [65, 66]. Human studies of the spinal analysis of the spinal cord and ventrolateral peri-
cord are limited. Structural neuroimaging of the aqueductal grey shows an increase in c-Fos
spine has shown early involvement of the spinal expression in streptozotocin model of neuropa-
cord in diabetic neuropathy, but no differences thy. In this model gabapentin reversed mechani-
were found between those with painful and pain- cal hypersensitivity and c-Fos expression [72].
less neuropathy [65, 67]. Thalamic changes are seen in human and ani-
The dorsal horn of the spinal cord is under the mal studies. Magnetic resonance spectroscopy
control of a descending pain modulatory system shows a reduction of N-acetyl-aspartate, a marker
that can either inhibit or facilitate transmission of for neuronal and axonal activity, in the thalamus
nociceptive information [68]. Once signals are of only those with painful diabetic neuropathy
transmitted along primary afferent neurones to [73]. Magnetic resonance perfusion shows an
the superficial dorsal horn, sensory information is increase in thalamic vascularity with associated
integrated by projection neurones in the superfi- sluggish flow in painful diabetic neuropathy [74].
cial and deep lamina of the spinal cord. This In streptozotocin models of diabetic neuropathy
information is then transmitted via ascending electrophysiological recordings of the ventropos-
pathways to the periaqueductal grey, rostroven- terolateral nucleus of thalamus show increased
tral medulla, and thalamus of the brain. The peri- spontaneous and evoked activity to mechanical
aqueductal grey and rostroventral medulla in turn stimulation of the hind paw [75]. Neurones in the
send projections back down to the spinal cord thalamic ventral posteromedial nucleus from rats
that can additionally facilitate or inhibit spinal with experimental diabetic neuropathy showed
excitability. Neuropathic pain can result from increased firing to precisely graded, multidirec-
signal amplification or loss of inhibition within tional whisker deflection, in the absence of overt
these central pathways. Conditioned pain modu- primary afferent dysfunction of trigeminal gan-
lation, a clinical measure of these descending glion neurones [76]. These findings suggest that
controls, is impaired in some patients with pain- central thalamic neurones are hyperexcitable in
ful diabetic neuropathy [69]. Functional brain the absence of increased or aberrant primary
imaging has shown that the ventrolateral periaq- afferent input.
ueductal grey functional connectivity is altered in Functional neuroimaging shows altered func-
those with painful diabetic polyneuropathy [70]. tional connectivity and somatosensory cortical
The magnitude of the changes correlates to spon- thickness in patients with painful diabetic neu-
taneous and evoked pain intensity. Therefore, ropathy [77–80]. For example, studies of the
dysfunction of the ventrolateral periaqueductal anterior cingulate cortex show increased func-
Pathophysiology of Neuropathic Pain 421

tional activity [81] and cerebral blood flow [82] peripheral sensory input and subsequently
in patients with painful diabetic neuropathy. In reduced cortical volumes and functional connec-
earlier studies, sensorimotor areas showed tivity. While in those with less severe neuropathy
reduced connectivity bilaterally, while several this phenomenon does not occur. It is not known
frontal areas, insulae, and thalami showed greater whether the CNS changes observed in painful
connectivity [77]. In more recent studies func- diabetic neuropathy are a maladaptive response
tional activity and somatosensory cortical thick- of increased peripheral nociceptive afferent input
ness are related to the sensory phenotype of the or an independent site for the initiation and main-
patient [83]. Patients with diabetic distal sym- tenance of neuropathic pain in diabetic
metrical neuropathy can be divided into two phe- neuropathy.
notypes based on quantitative sensory assessment
called irritable and non-irritable nociceptor/deaf-
ferentation phenotype. Irritable nociceptor phe- 4 Conclusions
notype presents with relatively preserved sensory
function associated with thermal and/or mechani- Over the last decade there have been significant
cal hyperalgesia, and the non-irritable nociceptor advances in our understanding of the pathophysi-
or deafferentation phenotype presents with sen- ological mechanisms of painful diabetic neuropa-
sory loss to thermal and mechanical stimuli. thy. The importance of voltage gated sodium
Current cohort studies show that the irritable channels and advances in structural and functional
nociceptor phenotype is far less common than the neuroimaging have provided novel insights into
non-irritable nociceptor phenotype [6, 8]. Patients peripheral and central mechanisms of painful dia-
with irritable nociceptor phenotype show greater betic neuropathy. It is clear from both clinical and
thalamus–insular cortex functional connectivity pre-clinical studies that an array of pathogenic
and decreased thalamus–somatosensory cortex mechanism underlies neuropathic pain and mul-
functional connectivity compared with those tiple mechanisms take place and are likely to con-
with the non-irritable nociceptor phenotype. tribute to some degree to the neuropathic pain
Thalamus–insular cortex functional connectivity experience on the level of an individual patient
was positively correlated to pain scores. Those (Tables 2 and 3, Fig. 1). What is needed are large
with a more severe polyneuropathy show a longitudinal studies utilising deep phenotyping
greater reduction in thalamus–somatosensory that will prospectively investigate the develop-
cortex functional connectivity and reduction in ment of painful diabetic neuropathy. Although
somatosensory grey matter cortical volume [83]. these studies may be logistically challenging and
This data suggests that distal axonopathy or expensive, these difficulties pale in contrast to the
peripheral deafferentation leads to reduced cost of painful diabetic neuropathy.
422 A. C. Themistocleous and M. M. Backonja

Cortex

PERCEPTION Thalamus

Brain
Stem
DECENDING
MODULARION

CENTRAL
TRANSMISSION Spinal Cord

K+/CL– cotransporter
Microglia

SYNAPTIC
Nav1.7, 1.8 TRANSMISSION
Cav3.2
TRANSDUCTION
PERIPHERAL
TRANSMISSION Dorsal Root
TRPA1 Ganglion (DRG)
TRPV1
Peripheral Nerves (PNS)

Pro-inflammatory cytokines

Fig. 1  Pain mechanisms represented along the neuroaxis

mation of severity, and treatments. Diabetes Care.


References 2010;33(10):2285–93.
5. Hébert HL, Veluchamy A, Torrance N, Smith
1. Finnerup NB, Haroutounian S, Kamerman P, Baron BH. Risk factors for neuropathic pain in diabetes mel-
R, Bennett DLH, Bouhassira D, et  al. Neuropathic litus. Pain. 2017;158(4):560–8.
pain: an updated grading system for research and 6. Raputova J, Srotova I, Vlckova E, Sommer C, Üçeyler
clinical practice. Pain. 2016;157(8):1599–606. N, Birklein F, et al. Sensory phenotype and risk fac-
2. von Hehn CA, Baron R, Woolf CJ. Deconstructing the tors for painful diabetic neuropathy: a cross-sectional
neuropathic pain phenotype to reveal neural mecha- observational study. Pain. 2017;158(12):2340–53.
nisms. Neuron. 2012;73(4):638–52. 7. Sieberg CB, Taras C, Gomaa A, Nickerson C, Wong
3. Abbott CA, Malik RA, van Ross ERE, Kulkarni C, Ward C, et  al. Neuropathic pain drives anxiety
J, Boulton AJM.  Prevalence and characteristics of behavior in mice, results consistent with anxiety
painful diabetic neuropathy in a large community-­ levels in diabetic neuropathy patients. Pain Rep.
based diabetic population in the UK. Diabetes Care. 2018;3(3):e651-e.
2011;34(10):2220–4. 8. Themistocleous AC, Ramirez JD, Shillo PR, Lees
4. Tesfaye S, Boulton AJM, Dyck PJ, Freeman R, JG, Selvarajah D, Orengo C, et al. The pain in neu-
Horowitz M, Kempler P, et  al. Diabetic neuropa- ropathy study (PiNS): a cross-sectional observational
thies: update on definitions, diagnostic criteria, esti- study determining the somatosensory phenotype
Pathophysiology of Neuropathic Pain 423

of painful and painless diabetic neuropathy. Pain. adult spinal sensory neurons, and is reexpressed fol-
2016;157(5):1132–45. lowing axotomy. J Neurophysiol. 1994;72(1):466–70.
9. Truini A, Spallone V, Morganti R, Tamburin S, 24. Bennett DL, Woods CG.  Painful and painless chan-
Zanette G, Schenone A, et al. A cross-sectional study nelopathies. Lancet Neurol. 2014;13(6):587–99.
investigating frequency and features of definitely 25. Craner MJ, Klein JP, Renganathan M, Black JA,
diagnosed diabetic painful polyneuropathy. Pain. Waxman SG.  Changes of sodium channel expres-
2018;159(12):2658–66. sion in experimental painful diabetic neuropathy. Ann
10. Calvo M, Davies AJ, Hébert HL, Weir GA, Chesler Neurol. 2002;52(6):786–92.
EJ, Finnerup NB, et  al. The genetics of neuropathic 26. Blesneac I, Themistocleous AC, Fratter C, Conrad LJ,
pain from model organisms to clinical application. Ramirez JD, Cox JJ, et al. Rare NaV1.7 variants asso-
Neuron. 2019;104(4):637–53. ciated with painful diabetic peripheral neuropathy.
11. Norbury TA, MacGregor AJ, Urwin J, Spector TD, Pain. 2018;159(3):469–80.
McMahon SB.  Heritability of responses to pain- 27. Han C, Themistocleous AC, Estacion M, Dib-
ful stimuli in women: a classical twin study. Brain. Hajj FB, Blesneac I, Macala L, et  al. The novel
2007;130(11):3041–9. activity of carbamazepine as an activation modu-
12. Scholz J, Rathmell JP, David WS, Chad DA, Broderick lator extends from Na(V)1.7 mutations to the
AC, Perros SG, et al. A standardized clinical evalua- Na(V)1.8-S242T Mutant Channel from a patient
tion of phenotypic diversity in diabetic polyneuropa- with painful diabetic neuropathy. Mol Pharmacol.
thy. Pain. 2016;157(10):2297–308. 2018;94(5):1256–69.
13. Demant DT, Lund K, Vollert J, Maier C, Segerdahl 28. Hoeijmakers JGJ, Faber CG, Merkies ISJ, Waxman
M, Finnerup NB, et  al. The effect of oxcarbaze- SG. Channelopathies, painful neuropathy, and diabe-
pine in peripheral neuropathic pain depends on tes: which way does the causal arrow point? Trends
pain phenotype: a randomised, double-blind, Mol Med. 2014;20(10):544–50.
placebo-­ controlled phenotype-stratified study. Pain. 29. Li QS, Cheng P, Favis R, Wickenden A, Romano
2014;155(11):2263–73. G, Wang H.  SCN9A variants may be implicated in
14. Jensen TS, Finnerup NB.  Allodynia and hyperalge- neuropathic pain associated with diabetic periph-
sia in neuropathic pain: clinical manifestations and eral neuropathy and pain severity. Clin J Pain.
mechanisms. Lancet Neurol. 2014;13(9):924–35. 2015;31(11):976–82.
15. Orstavik K, Jørum E.  Microneurographic findings 30. Estacion M, Vohra BPS, Liu S, Hoeijmakers J, Faber
of relevance to pain in patients with erythromelalgia CG, Merkies ISJ, et al. Ca2+ toxicity due to reverse
and patients with diabetic neuropathy. Neurosci Lett. Na+/Ca2+ exchange contributes to degeneration of
2010;470(3):180–4. neurites of DRG neurons induced by a neuropathy-­
16. Ørstavik K, Namer B, Schmidt R, Schmelz M, associated Nav1.7 mutation. J Neurophysiol.
Hilliges M, Weidner C, et  al. Abnormal function 2015;114(3):1554–64.
of C-Fibers in patients with diabetic neuropathy. J 31. Rolyan H, Liu S, Hoeijmakers JG, Faber CG, Merkies
Neurosci. 2006;26(44):11287–94. IS, Lauria G, et  al. A painful neuropathy-associated
17. Serra J, Duan WR, Locke C, Solà R, Liu W, Nothaft Nav1.7 mutant leads to time-dependent degeneration
W. Effects of a T-type calcium channel blocker, ABT-­ of small-diameter axons associated with intracellular
639, on spontaneous activity in C-nociceptors in Ca2+ dysregulation and decrease in ATP levels. Mol
patients with painful diabetic neuropathy: a random- Pain. 2016;12:1744806916674472.
ized controlled trial. Pain. 2015;156(11):2175–83. 32. Feldman EL, Nave K-A, Jensen TS, Bennett
18. Serra J, Bostock H, Solà R, Aleu J, García E, Cokic DLH.  New horizons in diabetic neuropathy:
B, et al. Microneurographic identification of sponta- mechanisms, bioenergetics, and pain. Neuron.
neous activity in C-nociceptors in neuropathic pain 2017;93(6):1296–313.
states in humans and rats. Pain. 2012;153(1):42–55. 33. Düll MM, Riegel K, Tappenbeck J, Ries V, Strupf
19. Serra J, Collado A, Solà R, Antonelli F, Torres X, M, Fleming T, et  al. Methylglyoxal causes pain and
Salgueiro M, et  al. Hyperexcitable C nociceptors in hyperalgesia in human through C-fiber activation.
fibromyalgia. Ann Neurol. 2014;75(2):196–208. Pain. 2019;160(11):2497–507.
20. Suzuki Y, Sato J, Kawanishi M, Mizumura K. Tissue 34. Bierhaus A, Fleming T, Stoyanov S, Leffler A, Babes
glucose level modulates the mechanical responses of A, Neacsu C, et  al. Methylglyoxal modification of
cutaneous nociceptors in streptozotocin-diabetic rats Nav1.8 facilitates nociceptive neuron firing and
but not normal rats in vitro. Pain. 2002;99(3):475–84. causes hyperalgesia in diabetic neuropathy. Nat Med.
21. Devor M.  Ectopic discharge in Abeta afferents 2012;18(6):926–33.
as a source of neuropathic pain. Exp Brain Res. 35. Andersson DA, Gentry C, Moss S, Bevan S. Transient
2009;196(1):115–28. receptor potential A1 is a sensory receptor for
22. Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-­ multiple products of oxidative stress. J Neurosci.
Hajj SD. The role of voltage-gated sodium channels 2008;28(10):2485–94.
in pain Signaling. Physiol Rev. 2019;99(2):1079–151. 36. Wei H, Hämäläinen Minna M, Saarnilehto M,
23. Waxman SG, Kocsis JD, Black JA. Type III sodium Koivisto A, Pertovaara A.  Attenuation of mechani-
channel mRNA is expressed in embryonic but not cal hypersensitivity by an antagonist of the TRPA1
424 A. C. Themistocleous and M. M. Backonja

Ion Channel in diabetic animals. Anesthesiology. streptozocin-induced diabetic neuropathy. Pain.


2009;111(1):147–54. 2009;145(1):184–95.
37. Andersson DA, Filipović MR, Gentry C, Eberhardt 51. Orestes P, Osuru HP, McIntire WE, Jacus MO,
M, Vastani N, Leffler A, et  al. Streptozotocin Salajegheh R, Jagodic MM, et  al. Reversal
stimulates the Ion Channel TRPA1 directly: of Neuropathic Pain in Diabetes by Targeting
INVOLVEMENT OF PEROXYNITRITE*. J Biol Glycosylation of Ca<sub>v</sub>3.2 T-Type
Chem. 2015;290(24):15185–96. Calcium Channels. Diabetes. 2013;62(11):3828–38.
38. Andersson DA, Gentry C, Light E, Vastani N, 52. Cameron NE, Eaton SE, Cotter MA, Tesfaye
Vallortigara J, Bierhaus A, et  al. Methylglyoxal S. Vascular factors and metabolic interactions in the
evokes pain by stimulating TRPA1. PLoS One. pathogenesis of diabetic neuropathy. Diabetologia.
2013;8(10):e77986. 2001;44(11):1973–88.
39. Huang J, Zhang X, McNaughton PA. Modulation of 53. Archer AG, Roberts VC, Watkins PJ. Blood flow pat-
temperature-sensitive TRP channels. Semin Cell Dev terns in painful diabetic neuropathy. Diabetologia.
Biol. 2006;17(6):638–45. 1984;27(6):563–7.
40. Hudson LJ, Bevan S, Wotherspoon G, Gentry C, Fox 54. Eaton SE, Harris ND, Ibrahim S, Patel KA, Selmi
A, Winter J.  VR1 protein expression increases in F, Radatz M, et  al. Increased sural nerve epineurial
undamaged DRG neurons after partial nerve injury. blood flow in human subjects with painful diabetic
Eur J Neurosci. 2001;13(11):2105–14. neuropathy. Diabetologia. 2003;46(7):934–9.
41. Kim HY, Park C-K, Cho I-H, Jung SJ, Kim JS, 55. Herder C, Kannenberg JM, Huth C, Carstensen-Kirberg
Oh SB.  Differential changes in TRPV1 expres- M, Rathmann W, Koenig W, et  al. Proinflammatory
sion after trigeminal sensory nerve injury. J Pain. cytokines predict the incidence and progression of dis-
2008;9(3):280–8. tal sensorimotor polyneuropathy: KORA F4/FF4 study.
42. Christoph T, Grünweller A, Mika J, Schäfer MKH, Diabetes Care. 2017;40(4):569–76.
Wade EJ, Weihe E, et al. Silencing of vanilloid recep- 56. Doupis J, Lyons TE, Wu S, Gnardellis C, Dinh T,
tor TRPV1 by RNAi reduces neuropathic and vis- Veves A.  Microvascular reactivity and inflamma-
ceral pain in  vivo. Biochem Biophys Res Commun. tory cytokines in painful and painless peripheral
2006;350(1):238–43. diabetic neuropathy. J Clin Endocrinol Metabol.
43. Ta LE, Bieber AJ, Carlton SM, Loprinzi CL, Low PA, 2009;94(6):2157–63.
Windebank AJ. Transient receptor potential Vanilloid 57. Purwata TE. High TNF-alpha plasma levels and mac-
1 is essential for cisplatin-induced heat hyperalgesia rophages iNOS and TNF-alpha expression as risk
in mice. Mol Pain. 2010;6:15. factors for painful diabetic neuropathy. J Pain Res.
44. Watabiki T, Kiso T, Kuramochi T, Yonezawa K, Tsuji 2011;4:169–75.
N, Kohara A, et al. Amelioration of neuropathic pain 58. Cheng HT, Dauch JR, Oh SS, Hayes JM, Hong Y,
by novel transient receptor potential Vanilloid 1 Feldman EL. p38 mediates mechanical allodynia
antagonist AS1928370 in rats without Hyperthermic in a mouse model of type 2 diabetes. Mol Pain.
effect. J Pharmacol Exp Ther. 2011;336(3):743–50. 2010;6:28.
45. Cui Y-Y, Xu H, Wu H-H, Qi J, Shi J, Li Y-Q. Spatio-­ 59. Zhang C, Ward J, Dauch JR, Tanzi RE, Cheng
temporal expression and functional involvement HT. Cytokine-mediated inflammation mediates pain-
of transient receptor potential Vanilloid 1  in dia- ful neuropathy from metabolic syndrome. PLoS One.
betic mechanical allodynia in rats. PLoS One. 2018;13(2):e0192333.
2014;9(7):e102052. 60. Tan AM, Samad OA, Fischer TZ, Zhao P, Persson
46. Pabbidi RM, Yu S-Q, Peng S, Khardori R, Pauza ME, A-K, Waxman SG.  Maladaptive dendritic spine
Premkumar LS.  Influence of TRPV1 on diabetes-­ Remodeling contributes to diabetic neuropathic pain.
induced alterations in thermal pain sensitivity. Mol J Neurosci. 2012;32(20):6795–807.
Pain. 2008;4:1744-8069-4-9. 61. Morgado C, Pereira-Terra P, Cruz CD, Tavares
47. Bestall SM, Hulse RP, Blackley Z, Swift M, Ved N, I. Minocycline completely reverses mechanical hyper-
Paton K, et al. Sensory neuronal sensitisation occurs algesia in diabetic rats through microglia-induced
through HMGB-1-RAGE and TRPV1 in high-glucose changes in the expression of the potassium chloride
conditions. J Cell Sci. 2018;131(14):jcs215939. co-transporter 2 (KCC2) at the spinal cord. Diabetes
48. Bourinet E, Francois A, Laffray S.  T-type calcium Obes Metab. 2011;13(2):150–9.
channels in neuropathic pain. Pain. 2016;157:S15–22. 62. Pabreja K, Dua K, Sharma S, Padi SS, Kulkarni
49. Joksimovic SL, Evans JG, McIntire WE, Orestes P, SK.  Minocycline attenuates the development of dia-
Barrett PQ, Jevtovic-Todorovic V, et al. Glycosylation betic neuropathic pain: possible anti-inflammatory
of ca(V)3.2 channels contributes to the hyperalgesia and anti-oxidant mechanisms. Eur J Pharmacol.
in peripheral neuropathy of type 1 diabetes. Front Cell 2011;661(1–3):15–21.
Neurosci. 2020;14:605312. 63. Tsuda M, Ueno H, Kataoka A, Tozaki-Saitoh H, Inoue
50. Messinger RB, Naik AK, Jagodic MM, Nelson K.  Activation of dorsal horn microglia contributes
MT, Lee WY, Choe WJ, et  al. In vivo silencing to diabetes-induced tactile allodynia via extracel-
of the CaV3.2 T-type calcium channels in sen- lular signal-regulated protein kinase signaling. Glia.
sory neurons alleviates hyperalgesia in rats with 2008;56(4):378–86.
Pathophysiology of Neuropathic Pain 425

64. Gold MS, Backonja MM.  Pain: from neurobiology p­ olyneuropathy: a clue to the pathogenesis of pain in
to disease. In: Zigmond MJ, Rowland LP, Coyle JT, type 1 diabetes. Diabetes Care. 2011;34(3):718–20.
editors. Neurobiology of brain disorders. San Diego: 75. Fischer TZ, Tan AM, Waxman SG.  Thalamic neu-
Academic Press; 2015. p. 674–92. ron hyperexcitability and enlarged receptive fields
65. Calcutt NA.  Diabetic neuropathy and neuropathic in the STZ model of diabetic pain. Brain Res.
pain: a (con)fusion of pathogenic mechanisms? Pain. 2009;1268:154–61.
2020;161(Suppl 1):S65–s86. 76. Freeman OJ, Evans MH, Cooper GJ, Petersen RS,
66. Zhou H-Y, Chen S-R, Pan H-L.  Targeting Gardiner NJ.  Thalamic amplification of sensory
N-methyl-­ D-aspartate receptors for treatment of input in experimental diabetes. Eur J Neurosci.
neuropathic pain. Expert Rev Clin Pharmacol. 2016;44(1):1779–86.
2011;4(3):379–88. 77. Cauda F, Sacco K, Duca S, Cocito D, D'Agata F,
67. Selvarajah D, Wilkinson ID, Emery CJ, Harris ND, Geminiani GC, et al. Altered resting state in diabetic
Shaw PJ, Witte DR, et  al. Early involvement of neuropathic pain. PLoS One. 2009;4(2):e4542.
the spinal cord in diabetic peripheral neuropathy. 78. Selvarajah D, Wilkinson ID, Fang F, Sankar A, Davies
Diabetes Care. 2006;29(12):2664–9. J, Boland E, et  al. Structural and functional abnor-
68. Kuner R, Kuner T. Cellular circuits in the brain and malities of the primary somatosensory cortex in dia-
their modulation in acute and chronic pain. Physiol betic peripheral neuropathy: a multimodal MRI study.
Rev. 2021;101(1):213–58. Diabetes. 2019;68(4):796–806.
69. Yarnitsky D, Granot M, Nahman-Averbuch H, 79. Teh K, Wilkinson ID, Heiberg-Gibbons F, Awadh
Khamaisi M, Granovsky Y. Conditioned pain modu- M, Kelsall A, Pallai S, et al. Somatosensory network
lation predicts duloxetine efficacy in painful diabetic functional connectivity differentiates clinical pain
neuropathy. Pain. 2012;153(6):1193–8. phenotypes in diabetic neuropathy. Diabetologia.
70. Segerdahl AR, Themistocleous AC, Fido D, Bennett 2021;64(6):1412–21.
DL, Tracey I. A brain-based pain facilitation mecha- 80. Wilkinson ID, Teh K, Heiberg-Gibbons F, Awadh M,
nism contributes to painful diabetic polyneuropathy. Kelsall A, Shillo P, et  al. Determinants of treatment
Brain. 2018;141(2):357–64. response in painful diabetic peripheral neuropathy: a
71. Silva M, Amorim D, Almeida A, Tavares I, Pinto-­ combined deep sensory phenotyping and multimodal
Ribeiro F, Morgado C. Pronociceptive changes in the brain MRI study. Diabetes. 2020;69(8):1804–14.
activity of rostroventromedial medulla (RVM) pain 81. Tseng MT, Chiang MC, Chao CC, Tseng WY, Hsieh
modulatory cells in the streptozotocin-diabetic rat. ST. fMRI evidence of degeneration-induced neuro-
Brain Res Bull. 2013;96:39–44. pathic pain in diabetes: enhanced limbic and striatal
72. Morgado C, Terra PP, Tavares I. Neuronal hyperactiv- activations. Hum Brain Mapp. 2013;34(10):2733–46.
ity at the spinal cord and periaqueductal grey during 82. Watanabe K, Hirano S, Kojima K, Nagashima K,
painful diabetic neuropathy: effects of gabapentin. Mukai H, Sato T, et  al. Altered cerebral blood flow
Eur J Pain. 2010;14(7):693–9. in the anterior cingulate cortex is associated with
73. Sorensen L, Siddall PJ, Trenell MI, Yue neuropathic pain. J Neurol Neurosurg Psychiatry.
DK.  Differences in metabolites in pain-processing 2018;89(10):1082–7.
brain regions in patients with diabetes and painful 83. Selvarajah D, Wilkinson ID, Maxwell M, Davies
neuropathy. Diabetes Care. 2008;31(5):980–1. J, Sankar A, Boland E, et  al. Magnetic resonance
74. Selvarajah D, Wilkinson ID, Gandhi R, Griffiths PD, neuroimaging study of brain structural differences
Tesfaye S.  Microvascular perfusion abnormalities in diabetic peripheral neuropathy. Diabetes Care.
of the thalamus in painful but not painless diabetic 2014;37(6):1681–8.
Central Nervous System
Involvement in Painful Diabetic
Neuropathy

Dinesh Selvarajah, Joyce Lim, Kevin Teh, Xin Chen,


Jing Wu, and Solomon Tesfaye

Abbreviations S2 Secondary somatosensory cortex


vlPAG Ventrolateral periaqueductal grey
1
H-MRS Proton magnetic resonance WM White matter
spectroscopy
ACC Anterior cingulate cortex
BOLD Blood oxygen level dependent
CNS Central nervous system 1 Painful Diabetic Neuropathy
DPN Diabetic peripheral neuropathy
DSP Diabetic sensorimotor polyneuropathy Painful distal symmetrical polyneuropathy (pain-
fMRI Functional magnetic resonance ful DPN) affects about 20% of people with diabe-
imaging tes [1] and is defined as ‘pain as a direct
GM Frey matter consequence of peripheral nerve injury’ [2]. It
HSMN Hereditary sensorimotor neuropathy often results in a significant morbidity and finan-
IR Irritable cial burden on the individual and a wider socio-
MR Magnetic resonance economic impact on society [3]. In the UK, the
NIR Non-irritable average annual healthcare cost per patient with
PAG Periaqueductal grey painful diabetic neuropathy is £2511 [3].
PET Positron emission tomography Approximately 60% of painful DPN patients
QST Quantitative sensory testing gainfully employed felt being less productive at
S1 Primary somatosensory cortex work [4]. Pain also interferes with their daily
activities, work, mood and sleep and is associated
D. Selvarajah · J. Lim · K. Teh with a reduced quality of life [4]. Not surpris-
Department of Oncology and Metabolism, Medical ingly, there is a high prevalence of depression
School, University of Sheffield, Sheffield, UK and emotional distress [4]. Hence, pain is a com-
X. Chen · J. Wu plex multidimensional construct which not only
Department of Endocrinology, Xiangya Hospital, affects sensory but also emotional/cognitive pro-
Central South University,
Changsha, People’s Republic of China cessing. Although nociceptive signals may be ini-
tiated/generated in the periphery, the central
S. Tesfaye (*)
Royal Hallamshire Hospital, Sheffield Teaching nervous system has an integral role in the patho-
Hospitals and the University of Sheffield, genesis and maintenance of the chronic pain
Sheffield, UK state/experience. Not surprisingly, the central
e-mail: Solomon.Tesfaye@sth.nhs.uk nervous system is the main site of action of many

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 427
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_24
428 D. Selvarajah et al.

commonly used pharmacotherapy agents in neu- involvement (see Fig.  1 below). One approach
ropathic pain. This further emphasises the impor- to assess this is by using quantitative sensory
tant role of the central nervous system in painful testing (QST) which has shown that painful
DPN [5–7]. DPN patients can be broadly subdivided into
Despite recent advances and considerable two phenotypes: ‘irritable’ (IR, with relatively
research, treatment for neuropathic pain is largely preserved sensory function associated with ther-
ineffective. A meta-analysis which evaluated the mal and/or mechanical hyperalgesia) and ‘non-
commonly used drugs in painful DPN found only irritable’ (NIR, dominated by thermal and
modest outcomes, with the number needed to mechanical sensory loss) [12]. Subsequent stud-
treat to achieve 50% pain relief ranging from 4 to ies appear to suggest that some treatments are
10 [8]. One reason for this poor treatment seemingly more effective in those with the irri-
response is that these agents do not target the table (IR) compared to the non-­irritable (NIR)
pathogenesis of neuropathic pain in diabetes. The nociceptor phenotype (for a review see [13] and
development of the current first-line agents fol- for examples see [14–17]). Consequently, pain
lowed a therapeutic pathway, rather than a molec- phenotyping may, in the future, become impor-
ular target pathway. Specifically, these agents tant in guiding individual patients’ treatment;
were developed for other indications before dem- although the exact approach is heavily under
onstration of analgesic efficacy in neuropathic debate. Whilst mechanistic approaches (e.g.
pain (e.g. pregabalin [anti-epileptic], tricyclic QST) that require carefully evaluating specific
antidepressants). Although the CNS binding site responses to guide therapy have significant
for these drugs has been identified (e.g. alpha-­ appeal (e.g. cold, heat, von Frey, etc.), in prac-
2delta-­1 calcium channel accessory protein for tice, these are time consuming and may be dif-
gabapentinoids) the mechanism whereby this ficult to implement in busy clinical practices.
interaction results in analgesia and the functional Furthermore, these are psychophysical measures
significance of this binding are still not clear. At which rely on patient responses and may be sub-
present, there is no pathogenetic treatment found jective and biased. Sensory profiling methods
to provide sufficient pain relief or prevent the also do not capture the complex and multidimen-
development of neuropathic pain in diabetes [9]. sional pain experience, which affects emotional
The main reason for this is because the pathogen- and cognitive processing in addition to sensory
esis of painful DPN is not fully understood. processing. For example, chronic pain patients
Given the importance of the central nervous sys- often undergo neuropsychological changes,
tem, a better understanding of the structural/func- which include changes in emotion and motiva-
tional neuroplastic changes that occur will serve tion or changes in cognition [18]. Chronic pain
as a catalyst for future drug discovery pro- may also arise after the onset of depression, even
grammes in painful DPN. This would also serve in patients without a prior history of pain or
as robust biomarkers capable of assessing the depression. Collectively, these clinical insights
benefits of future novel therapies [10]. suggest a better strategy is needed for assessing
Another reason for the wide variability or and treating painful DPN, given it is a chronic
poor treatment response seen in clinical practice disease of dynamic process (e.g. evolution of co-­
and the failure of numerous Phase III ran- morbid phenotypes such as anxiety or depres-
domised controlled trials of novel compounds sion), which is not easily reversed in most
for neuropathic pain is due to an underlying het- patients. This chapter will begin by examining
erogeneity in clinical pain phenotypes [11]. the alterations in the central nervous system
Painful DPN is not a homogenous condition but (CNS) in painful DPN and describe how neuro-
is comprised of many different clinical pain imaging could be used to phenotype patients with
phenotypes. This variation is thought to result painful DPN for personalised therapy before
from the extent and distribution of nerve fibre highlighting knowledge gaps and future work.
Central Nervous System Involvement in Painful Diabetic Neuropathy 429

Fig. 1  Schematic representation of the generation of sensitisation, grey star) that causes input from mechanore-
pain: (a) Normal: Central terminals of c-afferents project ceptive Aβ (light touch) and Aδ fibres (punctuate stimuli)
into the dorsal horn and make contact with secondary to be perceived as pain (allodynia). (c) C-fibre loss:
pain-signalling neurons. Mechanoreceptive Aβ afferents C-nociceptor degeneration and novel synaptic contacts of
project without synaptic transmission into the dorsal col- Aβ fibres with ‘free’ central nociceptive neurons, causing
umns (not shown) and also contact secondary afferent dynamic mechanical allodynia. (d) Central disinhibition:
dorsal horn neurons. (b) C-fibre sensitisation: Spontaneous Selective damage of cold-sensitive Aδ fibres that leads to
activity in peripheral nociceptors (peripheral sensitisation, central disinhibition, resulting in cold hyperalgesia.
black stars) induces changes in the central sensory pro- Sympat, sympathetic nerve [23]
cessing, leading to spinal cord hyperexcitability (central

2 CNS Changes in Response and the descending pain modulatory system can
to Peripheral Nerve Injury either facilitate or inhibit the transmission of
and How These Changes nociceptive information at spinal levels [22]. The
Result in Chronic Pain altered balance within these systems contributes
in Diabetes to the development and persistence of neuro-
pathic pain. A more detailed schematic represen-
Neuropathic pain is the result of maladaptive tation of the current theory for the generation of
changes in the peripheral and central nervous different symptoms and signs of neuropathic pain
systems [2, 19, 20]. Alterations in ion channels can be found in Fig. 1.
and ectopic activity at the periphery lead to
enhanced synaptic transmission within the dorsal
horn of the spinal cord and amplification of noci- 2.1 Changes in the Spinal Cord
ceptive information, a process known as central
sensitisation [21]. Central sensitisation and the Spinal cord involvement was first recognised in
altered balance between inhibitory interneurons the 1960s when post-mortem autopsy studies in
430 D. Selvarajah et al.

patients with advanced diabetes reported atrophy, a clear reduction in peripheral (pre- and postcen-
demyelination and axonal [24, 25] loss. These tral gyrus) and deep grey matter (GM) nuclei
findings were largely dismissed as being second- (caudate, putamen, medial pallidum, thalamus
ary to poor diabetes control and infection (e.g. and ventral nuclear) in regions involved with
syphilis) rather than DPN.  Indeed, the somatosensory/nociception perception in both
pathological abnormalities in the spinal cord
­ painful and painless DPN [30]. There were also
were reported in isolation and not examined in alterations in the spinothalamic, corticospinal
the context of DPN related peripheral nerve and thalamocortical white matter projections to
changes. Subsequent studies performed in the the cerebral cortex indicative of the involvement
late 1970s and 1980s utilised advances in somato- of somatosensory, motor and pain-related path-
sensory evoked potentials and demonstrated cen- ways [31]. Specific alterations in patients with
tral (brain and spinal cord) slowing in humans painful DPN have been reported in the cingulate
with DPN [26] and rodent models [27]. With the cortex, insular cortex, prefrontal lobe, thalamus,
advent and accessibility of magnetic resonance periaqueductal WM and external capsule [31].
(MR) imaging in the 1990s and early 2000s, The involvement of the anterior cingulate cortex
investigators were able to demonstrate clear spi- (ACC) and prefrontal cortex has been implicated
nal cord involvement in the form of cervical cord in the unpleasant perception associated with allo-
atrophy not only in patients with established dynia (pain due to a stimulus that normally does
DPN but also in those with early subclinical not provoke pain) [32]. Patients with painful- and
DPN. Eaton et al. revealed significant reduction painless DPN were also found to have reduced
in the spinal cord cross-sectional area in the cer- GM volume localised to regions involved in
vical and upper thoracic regions in patients with somatosensory perception compared to control
advanced DPN compared to non-diabetic con- patients with diabetes and no DPN and healthy
trols [28]. A subsequent, larger study confirmed volunteers [30]. Another modality of MR imag-
these findings but also revealed spinal cord atro- ing is proton (H+) MR spectroscopy. This method
phy was present in subjects with early, subclini- can be used to examine neurochemical altera-
cal DPN [29]. Moreover, subjects with hereditary tions in the brain. Using this method, patients
sensorimotor neuropathy (HSMN), an inherited with painless DPN had increased thalamic neuro-
neuropathy localised to the peripheral nerves, nal dysfunction compared to those with no DPN
had preserved cord cross-sectional area [29]. and painful DPN [33]. This finding suggests that
This argues against the ‘dying-back’ phenome- the preservation of thalamic neuronal function
non, i.e. peripheral nerve injury leading to subse- may be a pre-requisite for the perception of pain
quent cord atrophy in DPN rather it suggests that in diabetic neuropathy. This finding was also sup-
diabetes causes a generalised, concomitant insult ported by structural MR imaging which demon-
to both the peripheral and central nervous sys- strated reduction in thalamic grey matter volume
tems [29]. in subjects with painless diabetic neuropathy but
not painful diabetic neuropathy [30]. The thala-
mus plays an important role in somatosensory
2.2 Changes in the Brain perception. It not only acts as a relay station but
also modulates/processes nociceptive informa-
2.2.1 Structural/Metabolic tion from the periphery to other areas of the brain
Structural and functional cortical plasticity is a [34]. Animal models of painful DPN have dem-
fundamental property of the vertebrate central onstrated increased spontaneous activity from the
nervous system in response to nerve injury. thalamic nuclei [35]. Magnetic resonance perfu-
However, it can have maladaptive consequences, sion imaging of the brain has shown increased
possibly resulting in chronic pain. Studies using thalamic vascularity in patients with painful DPN
structural MR neuroimaging have demonstrated [36]. These findings imply the possible involve-
Central Nervous System Involvement in Painful Diabetic Neuropathy 431

ment of the thalamus in central amplification of ing pain in the lower limb region to include face
somatosensory signals. Similar changes have and lips regions [40] compared to painful DPN
also been observed in the ACC suggesting a role subjects with relatively preserved sensation.
for ACC activation in the development of central Furthermore, the extent to which S1 cortical
sensitisation [37]. In addition, patients with pain- structure and function are altered was related to
ful DPN have altered functional connectivity in the severity of neuropathy and the magnitude of
the ventrolateral PAG (vlPAG), which correlates self-reported pain. These data suggest a dynamic
with their pain intensity and cerebral blood flow plasticity of the brain in DPN, driven by the neu-
in response to tonic heat stimulation [38]. This ropathic process and may ultimately determine
supports the idea that abnormalities in the PAG an individual’s clinical pain phenotype [40].
may result not only in reduced inhibition, but also Over the last decade, resting-state fMRI
facilitation of pain [38]. Collectively, these find- (RS-fMRI), a relatively quick (6  min) and sim-
ings, supported by numerous studies in other ple, non-invasive technique, has become an
chronic pain conditions, demonstrate dynamic increasingly appealing method to examine spon-
neuronal changes that have profound effects on taneous brain activity in individuals without rely-
the brain in patients with DPN. ing on experimental external stimulation tasks.
During a typical RS-fMRI examination, the hae-
2.2.2 Functional modynamic response to spontaneous neuronal
Functional magnetic resonance imaging (fMRI) is activity (bold oxygen level dependent, BOLD
a modality of MR imaging which examines brain signal) is acquired, whilst subjects are instructed
activity by detecting changes in blood flow [blood to simply rest in the MRI scanner [41]. Data
oxygen level dependent (BOLD) signal] that is acquired is used to evaluate the connectivity
coupled with neuronal activation, i.e. when an area between different brain regions, i.e. functional
of the brain is in use, blood flow to that region also connectivity when a subject is at rest. RS-fMRI
increases and this can be examined using experiments in painful DPN have reported greater
fMRI. Using this method several studies have iden- thalamic-insula functional connectivity and
tified, the neurological signature for both physical decreased thalamic-somatosensory cortical func-
and neuropathic pain (capsaicin model). The brain tional connectivity in patients with the irritable
regions activated in response to a nociceptive stim- nociceptor phenotype compared to the non-­
ulus can be divided into somatic region responsible irritable nociceptor phenotype (Fig.  2) [42].
for the localisation of pain and coding intensity There was a significant positive correlation
such as the ventrolateral thalamus, somatosensory between thalamic-insula functional connectivity
cortex, dorsal posterior insula and the emotional/ with self-reported pain scores. Conversely, there
affective regions responsible for emotional pain was a greater reduction in thalamic-­somatosensory
processing such as the anterior insula, dorsal lateral cortical functional connectivity in those with
prefrontal cortex and ACC [39]. more severe neuropathy (Fig.  2). This demon-
A case-controlled, fMRI study of carefully strates how RS-fMRI measures of functional
phenotyped patients with DPN demonstrated a connectivity relate to both the somatic and non-­
pathophysiological relationship between anatom- somatic assessments of painful DPN.
ical and functional changes in the brain and clini- Multimodal MR imaging combining both
cal pain phenotypes. Subjects with painful DPN structural and RS-fMRI has also been used to
were divided into sensate (preserved nocicep- predict treatment response in painful DPN
tion) and insensate (abnormal nociception or (Fig.  2). Responders to neuropathic pain treat-
painful ‘painless DPN’) cohorts. Subjects with ment have significantly greater S1 cortical vol-
insensate painful DPN had the lowest S1 cortical ume and greater functional connectivity between
thickness and greatest S1 cortical functional reor- the insular cortex and corticolimbic system com-
ganisation with expansion of the area represent- pared to non-responders [43]. The insula cortex
432 D. Selvarajah et al.

Non Irritable Nociceptor Irritable Nociceptor

R Thal-Post CG FC
R PostCG R PostCG R Thal-lC FC
0.4
0.1

0.3

Effect size

Effect size
0.0
IC IC
0.2
Thalamus Thalamus
–0.1
0.1

–0.2 0
IR NIR IR NIR

NTSS-6 TCNS
F 1 2 3 4
0.6
Partial correlation (r)

0.4 * R Thal – IC FC

0.2 R Thal – PostCG FC


0
–0.2

*
–0.4

Responders Non-Responders

R IC-Forb FC R IC-Amy FC
0.5 0.4
Effect size

AC 0.3 AC

Effect size
0.2

IC IC
0.1
0.0
FOrb FOrb
Amy Amy
–0.1
Res N-Res Res N-Res

High IC to cortical limbic connectivity Low IC to cortical limbic connectivity

Fig. 2 (a) Right view of resting-state functional connec- FC, black). Bars 1–2 and 3–4 indicate correlations involv-
tivity in non-irritable nociceptor and irritable nociceptor ing the neuropathic total symptom score 6 (NTSS-6) and
painful diabetic peripheral neuropathy phenotype. R right, Toronto Clinical Neuropathy Score (TCNS), respectively.
IC insula cortex (MNI coordinates: 44, 4, 0), PostCG post- Each partial correlation coefficient, e.g. bar 1, is obtained
central gyrus (2–36 62), thal thalamus (10–19 6) [1]. (b, by correlating a given behaviour, e.g. self-reported pain
c) Bar charts demonstrating effect size of differences in scores (NTSS-6) with the functional connectivity derived
thalamic-insula and thalamic-somatosensory cortical from a given network, e.g. right thal-IC FC. *p < 0.05. FC
functional connectivity. IR irritable nociceptor, NIR non-­ functional connectivity [1]. (c) Right view of bilateral
irritable nociceptor. (b) The bar graph plots 4 FC and insular cortices resting-state FC in responders and non-­
behaviour partial correlation coefficients (r Pearson) responders to intravenous lidocaine treatment [2]. AC
derived from the functional connectivity of the right anterior cingulate gyrus, Acu nucleus accumbens, Amy
thalamus-­insula cortex (R thal-IC FC, white) and FC of amygdala, FOrb orbital frontal cortex, IC insula cortex, L
the right thalamus-somatosensory cortex (R thal-PostCG left, R right [42, 43]

plays a pivotal role in processing the emotion and 3 Clinical Implications


cognitive dimensions of the chronic pain experi-
ence [44]. The corticolimbic circuits have also There is now increasing evidence that brain
long been implicated in reward, decision making imaging could serve as a reliable biomarker in
and fear learning. Hence, these findings suggest preclinical and clinical trials in pain therapeutics
that this network may have a role in determining [13]. It could be used to increase the probability
treatment response in painful DPN.  Examining of novel compounds advancing to Phase II trials,
neurotransmitter activity and receptor ­distribution reduce the variability of the therapeutic response
within these networks may provide clues to why and reduce the overall expense and time of drug
some patients respond, whilst others are non- development. As highlighted in this chapter, MR
responders to neuropathic pain treatments. brain imaging has demonstrated alterations in
Central Nervous System Involvement in Painful Diabetic Neuropathy 433

brain structure, biochemistry and perfusion health economic analysis of screening and inter-
between painful and painless DPN. More recent ventions with study designs that allow relevant
studies have demonstrated significant structural and appropriate cost comparisons. Another cru-
and functional cortical changes linked not only to cial element is the patient’s perspective on the
different clinical phenotypes but also treatment usefulness, acceptance and feasibility of these
response in painful DPN. This has the potential novel imaging technologies in the provision of
to fast-track the development of new therapeutic future services.
interventions and alter the management approach
for painful DPN.  For example, MR imaging
could provide specific targets that are relevant to 4 Future Direction
pain and modulating activation in these targets
would provide objective evidence that a novel Future research can be divided into 3-key areas:
compound engages and attenuates nociceptive mechanistic or pathophysiological, performing
processing. clinical trials relevant to an individual with pain-
There are of course limitations and the useful- ful diabetic neuropathy and influencing federal
ness of biomarkers remains a controversial topic. agencies/commissioners of diabetes and pain ser-
Focused research is now needed to standardise vices. Brain imaging techniques such as func-
the application of neuroimaging biomarkers to tional magnetic resonance imaging (fMRI),
address the heterogeneity in their application in positron emission tomography (PET) and proton
terms of methodological approach, outcomes magnetic resonance spectroscopy (1H-MRS)
examined and time points used for assessments. provide neurochemical, structural or functional
More robust and better designed neuroimaging information on the processing and modulation of
studies are also required to develop and validate nociceptive inputs in the brain which result in the
central nervous system biomarkers capable of perception of pain [13]. A summary of the bene-
assessing the benefits of future novel therapies. fits and limitations of these different imaging
The direct clinical application of brain imaging modalities can be found in Table  1 [45–51].
in practice will also be dependent on a robust Neuroimaging studies have demonstrated differ-

Table 1  Some of the benefits and limitations of various neuroimaging modalities used in the investigation of CNS
lesions [46–52]
Neuroimaging
techniques Benefits Limitations
Functional • Detects local increased cerebral blood flow and • Low temporal resolution
MRI changes in blood oxygen concentration (BOLD • Signals may be affected by scanner’s
contrast) loud noise and physiologic noise
• Two primary types: Task-­based fMRI (utilisation of • Neurovascular uncoupling: False
visual, auditory or other stimuli to induce different negative BOLD responses due to
cognitive states in subjects) and resting-state fMRI interference of coupling between
(emphasis on spontaneous BOLD signal fluctuation) neuronal activity and adjacent
• Relatively high spatial resolution microvasculature
• Non-invasive
PET scan • Neuronal metabolic activity measured using • Involves the use of radioactive tracers
18
F-FDG can act as a marker of nerve injury and • Lower spatial resolution compared to
neuropathic pain fMRI
• Non-invasive
1
H-MRS • Non-invasive • Low sensitivity
• Does not involve ionising radiation • Lack of standardisation and quality
• Able to investigate a wide range of biological assurance of data acquisition and
processes analysis methods
• Combination of spectroscopy and imaging enables
the acquisition of metabolic, physiological and
anatomical data in a single experiment
434 D. Selvarajah et al.

ences in neuronal activation in response to acute n­ ociceptive inputs will complete the picture of
pain and chronic pain as well as differences in CNS modulation of neuropathic pain. This poses
functional connectivity between patients and an enormous imaging challenge because the
healthy controls. This provides an important small size of the spinal cord results in low signal
segue into a better understanding of the underly- to noise and there is considerable motion artefact
ing mechanisms involved in the generation, from respiration, cardiac cycle and cerebrospinal
maintenance and exacerbation of chronic pain fluid pulsations [53]. These challenges are now
[13]. As a prognostic tool, brain imaging could being addressed with high-field strength MR
also be used to identify individuals who are more imaging at 7 and 14-tesla opening up this part of
‘vulnerable’ or ‘resilient’ against developing per- the neural axis for future mechanistic studies and
sistent pain after being exposed to factors which clinical applications [54, 55].
can trigger the onset of chronic pain. For exam- Another promising imaging innovation that
ple, increased functional connectivity of the will significantly impact clinical and experi-
nucleus accumbens with prefrontal cortex pre- mental pain research is simultaneous positron
dicts transition from acute pain to chronic pain in emission tomography and MR imaging (PET/
subjects with sub-acute back pain [52]. Similar MR). This is a powerful non-invasive tool with
studies need to be performed in painful DPN to strong translational value. PET imaging serves
determine if the same or other brain regions are as a bridge from in  vitro pharmacology to
implicated. Brain imaging can also be used to in vivo molecular readouts, whereas fMRI is an
predict treatment response, hence may enable indicator of dynamic brain function with a close
stratification of participants in clinical trials [13]. link to behaviour (Fig. 3). The PET radiotracer
Proof of concept for this has been demonstrated 2-[18F] fluoro-2-deoxy-d-glucose ([18F]FDG)
in fibromyalgia where fMRI data derived from is able to quantify not only changes in brain
visual stimulation has shown a greater than 80% activity that is reflected by increases and
accuracy in distinguishing whether a patient with decreases in cerebral metabolic rate of glucose
fibromyalgia improved when administered pre- but also the brain activity that stems primarily
gabalin vs. placebo [53]. In painful DPN, a quick from synaptic activity [56]. Combining [18F]
(6-min) RS-fMRI scan could also be used to FDG-PET with fMRI allows the study of neu-
determine response to neuropathic pain treatment ronal activity on a slow time scale (minutes)
[42]. Hence, brain imaging can be a useful diag- with metabolic-level PET data, and on a fast
nostic, prognostic and predictive tool and its use time scale (seconds) with vascular-­level fMRI
as a potential biomarker in chronic pain clinical data (Fig.  2) [56]. Thus, PET-MR provides a
trials requires further research focussing on stan- more robust, direct readout of neuronal activity
dardisation of outcomes, validation, reproduc- in-vivo. It represents a key bridging technology,
ibility and evaluation of outcomes. whereby functional markers with molecular
The next important step in delineating CNS and/or pharmacological specificity can be
involvement in diabetic neuropathy is to develop quantified and tracked with anatomical preci-
methods that allow non-invasive assessment of sion. Combined with advanced computational
the functional changes in the spinal cord in-vivo. models, PET/MR provides a level of insight
This will be an important step in exploring inter-­ into disease processes and drug effects in vivo
relationships between the peripheral and central which corresponds much more closely to what
nervous system in greater detail. As detailed is possible in preclinical research. Another key
above, there have been a number of anatomical advance is the availability of PET radio-­ligands
studies which have demonstrated increased cord for opioid [57] and dopamine receptors [58].
atrophy in diabetic neuropathy—however, accu- This has allowed the study of these receptor
rate assessment of (1) neuronal activity within systems in a number of clinical pain states but
the spinal cord in relation to evoked/spontaneous presently remains solely a research tool.
pain and (2) top-down inhibition of afferent Preliminary studies have showed abnormal
Central Nervous System Involvement in Painful Diabetic Neuropathy 435

Neurophysiological Haemodynamic
Neuropharmacological Neurometabolic
Glycogen
HO CH2CHCO2H

HO NH2 Glucose Glucose


NO

Pyruvate K+
Stimulation
CO2
Protein Fat
O2
TCA
Energy cycle Nutrients

Pharmacon- Increase in
Increase energy Increase in
receptor neuronal
consumption blood flow
interaction activity

PET MEG, EEG PET fMRI


Imaging

Receptor FMRI Lorenz Glucose Blood Flow Blood


Occupation effect imaging consumption oxygenation (BOLD)
imaging diffusion

Fig. 3  An illustration of the different specificity of func- lism and haemodynamics. PET positron emission
tional neuroimaging modalities to physiological processes tomography, MEG magnetoencephalography, EEG elec-
(receptor activity, electromagnetic, metabolic and haemo- troencephalography, fMRI functional magnetic resonance
dynamic) involved in neuronal activity, cerebral metabo- imaging [61]

receptor binding in a number of different with the publication of various consensus state-
­neuropathic pain states [59] and reversal of this ments [61, 62]. Emerging approaches to imaging
abnormality following the resolution of painful analyses such as machine learning algorithms
symptoms [60]. The main limitation with many (e.g. support vector machines, convolutional neu-
of these cross-sectional, observational studies ral networks, etc.) may also prove useful in
is whether the changes described are the cause addressing concerns about replication [13, 42].
or consequence of the chronic pain state. These methods enable pooling of MR data from
However, it is evident from studies in painful studies utilising different imaging equipment and
diabetic neuropathy that the brain is ailing both analysis procedures and have produced informa-
in terms of functional reorganisation and exten- tive inferences about brain structure and function
sive neurodegeneration. in diabetic neuropathy. These are essential steps
Increase assay sensitivity in clinical trial— towards gaining regulatory approvals from fed-
exposing fewer patients to risk of new drugs and eral agencies for the use of neuroimaging as a
reducing costs of Phase 3 studies. Another impor- validated biomarker in diabetic painful DPN
tant aspect of neuroimaging research especially [13]. This would lead to increased assay sensitiv-
in the context of chronic pain trials is the need to ity in later stage clinical trials (e.g. phase 2b or 3)
standardise the use of neuroimaging tools and studies which will minimise patient exposure to
post-processing methods. Significant steps have experimental drugs and has the potential to
been made towards addressing this limitation reduce the overall costs of drug development.
436 D. Selvarajah et al.

Disease modifying treatments that target the ies described have been relatively small, cohort
pathogenesis of diabetic neuropathy and effective studies that were exploratory that could lead to
symptoms control remain the highest unmet false positive results as a consequence of an
needs. As of yet, standard recommendations to inflated risk of type 1 error. Nevertheless, neuro-
optimise cardiovascular risk factors effectively imaging led developments for people with diabe-
(in some countries where there is local regulatory tes will have wider implications for other chronic
approval for the use of agents such as aldose pain aetiologies outside the field of diabetes and
reductase inhibitors, alpha-lipoic acid and benfo- hence are highly relevant.
tiamine) and symptom control are the corner-
stones in practice [63]. In the future, these
recommendations could be augmented by per- References
sonalised, targeted approaches towards restoring
neurotransmitter imbalance, cortical neuronal 1. Veves A, Backonja M, Malik RA. Painful diabetic neu-
ropathy: epidemiology, natural history, early diagno-
degeneration/functional reorganisation and fur- sis, and treatment options. Pain Med. 2008;9:660–74.
ther potential factors addressed in this chapter 2. Sloan G, et al. A new look at painful diabetic neuropa-
that might represent treatment targets. As in other thy. Diabetes Res Clin Pract. 2018;144:177–91.
microvascular complications for diabetes, e.g. 3. Alleman CJM, et  al. Humanistic and economic bur-
den of painful diabetic peripheral neuropathy in
diabetic nephropathy with microalbuminuria or Europe: a review of the literature. Diabetes Res Clin
retinopathy with digital retinal photography, the Pract. 2015;109:215–25.
identification of a robust biomarker for painful 4. Selvarajah D, et  al. The contributors of emotional
DPN will lead to early identification and also distress in painful diabetic neuropathy. Diab Vasc Dis
Res. 2014;11:218–25.
accelerate the development of targets of the spe- 5. Feldman EL, et al. Diabetic neuropathy. Nat Rev Dis
cific mechanism that leads to chronic pain. Primers. 2019;5(1):41.
Moreover, the disease processes that are not 6. Alam U, Sloan G, Tesfaye S.  Treating pain in dia-
unique to diabetes but also contribute to the per- betic neuropathy: current and developmental drugs.
Drugs. 2020;80(4):363–84. https://doi.org/10.1007/
sistence and resistance to treatment of the chronic s40265-­020-­01259-­2.
pain condition will be elucidated further. For 7. Singh R, Rao HK, Singh TG.  Neuropathic pain in
example, the CNS processes described in this diabetes mellitus: challenges and future trends. Obes
chapter in patients with painful DPN are proba- Med. 2020;18:100215.
8. Finnerup NB, et al. Pharmacotherapy for neuropathic
bly still important contributors to chronic pain in pain in adults: systematic review, meta-analysis and
people without diabetes. updated NeuPSig recommendations. Lancet Neurol.
2015;14:162–73.
9. Kioskli K, Scott W, Winkley K, Kylakos S, McCracken
LM. Psychosocial factors in painful diabetic neuropa-
5 Conclusions thy: a systematic review of treatment trials and survey
studies. Pain Med. 2019;20:1756–73.
In conclusion, clinicians and patients acknowl- 10. Bönhof GJ, et  al. Emerging biomarkers, tools, and
edge that painful DPN is an important complica- treatments for diabetic polyneuropathy. Endocr Rev.
2018;40:153–92.
tion of diabetes that leads to significant 11. Finnerup NB, Sindrup SH, Jensen TS. The evidence
comorbidity and suffering. Hitherto considered a for pharmacological treatment of neuropathic pain.
disease of the peripheral nervous system, research Pain. 2010;150:573–81.
over the past decades has outlined the clinical 12. Backonja MM, et  al. Value of quantitative sensory
testing in neurological and pain disorders: NeuPSIG
features and brain/spinal cord imaging correlates consensus. Pain. 2013;154:1807–19.
of painful DPN.  Insights gained from clinical 13. Smith SM, Dworkin RH, Turk DC, Baron R,
research are beginning to be translated into appli- Polydefkis M, Tracey I, Borsook D, Edwards RR,
cations but there are still gaps in our knowledge. Harris RE, Wager TD, Arendt-Nielsen L. The poten-
tial role of sensory testing, skin biopsy, and func-
Current challenges include replication to provide tional brain imaging as biomarkers in chronic pain
further support for the neuroimaging tools. It is clinical trials: IMMPACT considerations. J Pain.
important to acknowledge that most of the stud- 2017;18:757–77.
Central Nervous System Involvement in Painful Diabetic Neuropathy 437

14. Mainka T, et  al. Presence of hyperalgesia predicts 29. Selvarajah D, et  al. Early involvement of the spinal
analgesic efficacy of topically applied capsaicin 8% in cord in diabetic peripheral neuropathy. Diabetes Care.
patients with peripheral neuropathic pain. Eur J Pain. 2006;29:2664–9.
2016;20:116–29. 30. Selvarajah D, Wilkinson ID, Maxwell M, Davies
15. Edwards RR, et al. Patient phenotyping in clinical tri- J, Sankar A, Boland E, et  al. Magnetic resonance
als of chronic pain treatments: IMMPACT recommen- neuroimaging study of brain structural differences
dations. Pain. 2016;157:1851–71. in diabetic peripheral neuropathy. Diabetes Care.
16. Wasner G, Kleinert A, Binder A, Schattschneider J, 2014;37(6):1681–8.
Baron R.  Postherpetic neuralgia: topical lidocaine 31. Zhang Y, et al. Sensorimotor and pain-related altera-
is effective in nociceptor-deprived skin. J Neurol. tions of the gray matter and white matter in type 2 dia-
2005;252:677–86. betic patients with peripheral neuropathy. Hum Brain
17. Demant DT, Lund K, Vollert J, Maier C, Segerdahl M, Mapp. 2020;41:710–25.
Finnerup NB, Jensen TS, Sindrup SH. The effect of 32. Jensen TS, Finnerup NB.  Allodynia and hyperalge-
oxcarbazepine in peripheral neuropathic pain depends sia in neuropathic pain: clinical manifestations and
on pain phenotype: a randomised, double-blind, mechanisms. Lancet Neurol. 2014;13:924–35.
placebo-controlled phenotype-stratified study. Pain. 33. Selvarajah D, Wilkinson ID, Emery CJ, Shaw PJ,
2014;155(11):2263–73. Griffiths PD, Gandhi R, Tesfaye S. Thalamic neuronal
18. Tracey I. “Seeing” how our drugs work brings dysfunction and chronic sensorimotor distal symmet-
translational added value. Anesthesiology. rical polyneuropathy in patients with type 1 diabetes
2013;119(6):1247–8. mellitus. Diabetologia. 2008;51(11):2088–92.
19. Jensen TS, Finnerup NB.  Neuropathic pain: periph- 34. Shillo P, et  al. Painful and painless diabetic neu-
eral and central mechanisms. Eur J Pain Suppl. ropathies: what is the difference? Curr Diab Rep.
2009;3:33–6. 2019;19:32.
20. Meacham K, Shepherd A, Mohapatra DP, 35. Freeman OJ, Evans MH, Cooper GJS, Petersen
Haroutounian S.  Neuropathic pain: central vs. RS, Gardiner NJ.  Thalamic amplification of sen-
peripheral mechanisms. Curr Pain Headache Rep. sory input in experimental diabetes. Eur J Neurosci.
2017;21:28. 2016;44:1779–86.
21. Feldman EL, Nave KA, Jensen TS, Bennett DLH. New 36. Selvarajah D, Wilkinson ID, Gandhi R, Griffiths
horizons in diabetic neuropathy: mechanisms, bioen- PD, Tesfaye S.  Microvascular perfusion abnor-
ergetics, and pain. Neuron. 2017;93:1296–313. malities of the thalamus in painful but not painless
22. Colloca L, et  al. Neuropathic pain. Nat Rev Dis diabetic polyneuropathy: a clue to the pathogen-
Primers. 2017;3:17002. esis of pain in type 1 diabetes. Diabetes Care.
23. Vinik A, Casellini C, Nevoret ML.  Diabetic neu- 2011;34(3):718–20.
ropathies. In: Feingold KR, Anawalt B, Boyce 37. Watanabe K, et  al. Altered cerebral blood flow in
A, et  al., editors. Endotext [Internet]. South the anterior cingulate cortex is associated with
Dartmouth, MA: MDText.com; 2000. https:// neuropathic pain. J Neurol Neurosurg Psychiatry.
www.ncbi.nlm.nih.gov/books/NBK279175/figure/ 2018;89:1082–7.
diab-­neuropathies_f_diab-­neuropathies_figure8/. 38. Segerdahl AR, Themistocleous AC, Fido D, Bennett
24. Reske-Nielsen E, Lundbaek K.  Pathological DL, Tracey I. A brain-based pain facilitation mecha-
changes in the central and peripheral nervous nism contributes to painful diabetic polyneuropathy.
system of young long-term diabetics. II.  The spi- Brain. 2018;141:357–64.
nal cord and peripheral nerves. Diabetologia. 39. Wager TD, et  al. An fMRI-based neurologic
1968;4:34–43. signature of physical pain. N Engl J Med.
25. Reske-Nielsen E, Lundbaek K, Gregersen G, Harmsen 2013;368:1388–97.
A. Pathological changes in the central and peripheral 40. Selvarajah D, et  al. Structural and functional abnor-
nervous system of young long-term diabetics: the malities of the primary somatosensory cortex in dia-
terminal neuro-muscular apparatus. Diabetologia. betic peripheral neuropathy: a multimodal MRI study.
1970;6:98–103. Diabetes. 2019;68:796–806.
26. Ziegler D, Mühlen H, Dannehl K, Gries FA.  Tibial 41. Lv H, et al. Resting-state functional MRI: everything
nerve somatosensory evoked potentials at various that nonexperts have always wanted to know. Am J
stages of peripheral neuropathy in insulin dependent Neuroradiol. 2018;39:1390–9.
diabetic patients. J Neurol Neurosurg Psychiatry. 42. Teh K, Wilkinson ID, Heiberg-Gibbons F, Awadh M,
1993;56(1):58–64. Kelsall A, Pallai S, Sloan G, Tesfaye S, Selvarajah
27. Biessels GJ, et  al. Neurophysiological changes D.  Somatosensory network functional connectivity
in the central and peripheral nervous system of differentiates clinical pain phenotypes in diabetic neu-
streptozotocin-­diabetic rats. Course of development ropathy. Diabetologia. 2021;64(6):1412–21.
and effects of insulin treatment. Brain. 1999;122(Pt 43. Wilkinson ID, et  al. Determinants of treatment
4):757–68. response in painful diabetic peripheral neuropathy: a
28. Eaton SEM, et al. Spinal-cord involvement in diabetic combined deep sensory phenotyping and multimodal
peripheral neuropathy. Lancet. 2001;358:35–6. brain MRI study. Diabetes. 2020;69:1804–14.
438 D. Selvarajah et al.

44. Lu C, et al. Insular cortex is critical for the perception, 55. Maieron M, et al. Functional responses in the human
modulation, and Chronification of pain. Neurosci spinal cord during willed motor actions: evidence
Bull. 2016;32:191–201. for side- and rate-dependent activity. J Neurosci.
45. Glover GH.  Overview of functional magnetic reso- 2007;27:4182–90.
nance imaging. Neurosurg Clin N Am. 2011;22:133–9. 56. Wehrl HF, Hossain M, Lankes K, Liu CC, Bezrukov
46. Chen JE, Glover GH.  Functional magnetic reso- I, Martirosian P, Schick F, Reischl G, Pichler
nance imaging methods. Neuropsychol Rev. BJ.  Simultaneous PET-MRI reveals brain function
2015;25:289–313. in activated and resting state on metabolic, hemo-
47. Shruti A, Sair HI, Pillai JJ. Limitations of rsfMRI in dynamic and multiple temporal scales. Nat Med.
the setting of focal brain lesions. Neuroimaging Clin 2013;19:1184–9.
N Am. 2017;27:645–61. 57. Jones AKP, et  al. Measurement of changes in opi-
48. Slough C, Masters SC, Hurley RA, Taber KH. Clinical oid receptor binding in  vivo during trigeminal neu-
positron emission tomography (PET) neuroimag- ralgic pain using [11C]diprenorphine and positron
ing: advantages and limitations as a diagnostic tool. J emission tomography. J Cereb Blood Flow Metab.
Neuropsychiatry Clin Neurosci. 2016;28:1–6. 1999;19:803–8.
49. Behera D, Jacobs KE, Behera S, Rosenberg J, Biswal 58. Hagelberg N, et  al. Striatal dopamine D2 receptors
S. 18F-FDG PET/MRI can be used to identify injured in modulation of pain in humans: a review. Eur J
peripheral nerves in a model of neuropathic pain. J Pharmacol. 2004;500:187–92.
Nucl Med. 2011;52:1308–12. 59. Harris RE, et  al. Decreased central μ-opioid
50. Chatham JC, Blackband SJ.  Nuclear magnetic reso- receptor availability in fibromyalgia. J Neurosci.
nance spectroscopy and imaging in animal research. 2007;27:10000–6.
ILAR J. 2001;42:189–208. 60. Jones AKP, et  al. Changes in central opioid recep-
51. Öz G, et  al. Clinical proton MR spectroscopy tor binding in relation to inflammation and pain in
in central nervous system disorders. Radiology. patients with rheumatoid arthritis. Rheumatology.
2014;270:658–79. 1994;33:909–16.
52. Baliki MN, et  al. Corticostriatal functional connec- 61. Uludağ K, Roebroeck A.  General overview on the
tivity predicts transition to chronic back pain. Nat merits of multimodal neuroimaging data fusion.
Neurosci. 2012;15:1117–9. Neuroimage. 2014;102:3–10.
53. López-Solà M, Woo CW, Pujol J, Deus J, Harrison 62. Huang L, et al. Brain white matter changes associated
BJ, Monfort J, Wager TD.  Towards a neuro- with urological chronic pelvic pain syndrome: multi-
physiological signature for fibromyalgia. Pain. site neuroimaging from a MAPP case-control study.
2017;158(1):34–47. Pain. 2016;157:2782–91.
54. Brooks JCW, et al. Physiological noise modelling for 63. Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diag-
spinal functional magnetic resonance imaging stud- nosis and clinical management of diabetic peripheral
ies. Neuroimage. 2008;39:680–92. neuropathy. Nat Rev Endocrinol. 2021;17(7):400–20.
Part III
Clinical Consequences and Treatments
Characteristics and Treatment
of Painful Diabetic Neuropathy

Sandra Sif Gylfadottir and Nanna Brix Finnerup

1 Definition, Epidemiology, caused by a lesion or disease of the somatosen-


Diagnosis, sory nervous system” [4]. Of patients with DPN,
and Characteristics 25–50% experience pain due to their DPN [5–
11]. Many factors influence the prevalence esti-
Painful diabetic neuropathies include painful dia- mates of P-DPN, including different definitions
betic radiculoplexopathy, mononeuropathies, and assessment methods of P-DPN as well as dif-
treatment-induced neuropathies induced by fast ferences in duration and type of diabetes [8,
improvement of glycemic control after hypergly- 12–19].
cemia, entrapment neuropathies, and distal sym- Patients with P-DPN often complain of both
metrical sensorimotor polyneuropathies [1]. The painful and non-painful sensations. Non-painful
focus of this chapter will be on painful diabetic sensations include paresthesia, which are not
polyneuropathy (P-DPN) as this group represents unpleasant, e.g. tingling, and dysesthesia, which
the majority of the patients in clinical setting [2] are unpleasant sensations, e.g. pricking. The pain
and the diagnostic and treatment approaches to can be spontaneous or evoked and examples of
the different types of painful diabetic neuropa- neuropathic pain symptom descriptions can be
thies are similar, except for treatment-induced burning, squeezing, shooting, or pricking pain,
neuropathy (discussed in chapter “Asymmetric although the pain can be described in a similar
Diabetic Neuropathy: Radiculoplexus way as in other pain conditions [20]. Therefore
Neuropathies, Mononeuropathies, and Cranial the pain description itself is normally not suffi-
Neuropathies”). cient to diagnose P-DPN and it has to be con-
Pain is defined as “An unpleasant sensory and firmed by a clinical examination and supportive
emotional experience associated with, or resem- diagnostic tests. Evoked pain to, e.g. touch or
bling that associated with, actual or potential tis- cold is uncommon in P-DPN compared to some
sue damage” [3]. P-DPN is a subgroup of other neuropathic pain conditions. Pain and other
neuropathic pain, which is defined as “pain abnormal sensory symptoms usually start in the
feet and may spread to the legs and hands in the
S. S. Gylfadottir (*) · N. B. Finnerup most severe cases.
Department of Clinical Medicine, Danish Pain The clinical examination consists of a stan-
Research Center, Aarhus University, dard neurological examination with a focus on
Aarhus, Denmark sensory examination of feet and hands, or any
Department of Neurology, Aarhus University other affected area. In Fig. 1 there are examples
Hospital, Aarhus, Denmark of tools that can be used in the bedside examina-
e-mail: sifgyl@clin.au.dk; finnerup@clin.au.dk

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 441
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_25
442 S. S. Gylfadottir and N. B. Finnerup

tion of a patient presenting with P-DPN. In 2016 clinical practice [21, 22]. Applying this for
the International Association for the Study of P-DPN (Fig. 2), possible P-DPN requires a his-
pain special interest group on neuropathic pain tory of P-DPN, pain in the feet, and a diagnosis of
(NeuPSIG) proposed an upgraded grading sys- diabetes. Probable P-DPN additionally requires
tem for neuropathic pain, both for research and sensory signs in the same area, and definite
P-DPN requires abnormal tests that confirm a
structural or functional damage to the sensory
nerves (examples: skin biopsy and nerve conduc-
tion studies). At all stages it is important to con-
sider other causes of polyneuropathy with similar
or the same symptom presentation (i.e. alcohol or
chemotherapy induced, infective or inflammatory
polyneuropathy) or conditions presenting with
painful feet or legs (i.e. arthritis, spinal stenosis,
herniated disc, peripheral arterial diseases) [10].
It is difficult to replace bedside clinical exami-
nation with standardized assessment tools,
although they can be useful in screening, epide-
miological research, and to aid in the diagnosis of
neuropathic pain [23]. An example of screening
and diagnostic tool is the douleur neuropathique
en 4 questions (DN4). The DN4 consists of seven
Fig. 1  Example of tools that can be used in the bedside
examination of the patient presenting with P-DPN. Left to questions and a clinical examination part and was
right: tuning fork for vibration sense, brush for light brush designed to discriminate neuropathic pain from
stroking, Neuropen® for light touch and pinprick sensa- other types of pain [24]. The questions are focused
tion, reflex hammer, temperature rolls, cotton wool swab, on pain and paresthesia (pins and needles, numb-
and ear pin

Painful diabetic polyneuropathy

Possible painful DPN

Diabetes and pain in both feet

Other causes of pain


excluded

Probable painful DPN

Pain is associated with sensory signs in the feet/


legs, neuroanatomically plausible with a
polyneuropathy

Definite painful DPN

Abnormal NCS and/or IENFD

Fig. 2  The diagnosis of P-DPN [21]. At all steps, other causes of pain and neuropathy should be considered
Characteristics and Treatment of Painful Diabetic Neuropathy 443

ness, itching, burning, painful cold, and electric with P-DPN and pain intensity [7, 12, 17, 31, 32,
shock sensation) and the examination of brush 35–40]. Most studies are cross-sectional and
and pinprick hypoesthesia and dynamic mechani- have therefore not examined the causal relation-
cal allodynia (DMA) in the feet/legs. The DN4 ship between having a chronic neuropathic pain
questionnaire part has been validated and used for and mental health. A large questionnaire study of
P-DPN [19, 24, 25]. In a recent validation study in Danish patients with type 2 diabetes and neurop-
type 2 diabetes patients, the sensitivity and speci- athy found that both patients with DPN with and
ficity of the DN4 questionnaire for definitely without pain reported worse mental health and
diagnosed P-DPN were 80.0 (44.4; 97.5) and 89.9 quality of life, although patients with pain were
(83.6; 94.3), ­respectively, for a cut of point ≥3/7 more severely affected [38].
[10]. An example of a questionnaire that can be
used to quantify and characterize the symptoms of
neuropathic pain is the neuropathic pain symptom 2 Treatment of P-DPN
inventory (NPSI) with 12 questions about the
presence, character, and intensity of pain and the Information about pain and pain mechanisms and
answers can be converted into sum scores [26]. discussion of aggravating and alleviating factors
Other neuropathic pain tools are available, such as and pain coping strategies are often the first step
the PainDETECT, the Leeds Assessment of in pain treatment. While the evidence for psycho-
Neuropathic Symptoms and Signs questionnaire, logical and self-care interventions are limited for
and the Neuropathic Pain Questionnaire [23]. The chronic pain, most guidelines recommend this in
numerical rating scale (NRS) from 0 to 10 or a the treatment of chronic pain [41]. For any treat-
Visual Analog Scale (VAS) is used to measure the ment, realistic expectations for the outcome
intensity of the pain with 0 being no pain and ten should be discussed with the patient, as often
the worst pain imaginable and is simple to admin- only partial pain relief can be expected. A multi-
ister [27]. In the assessment of pain it is also disciplinary approach with physicians, pain
important to assess the impact of pain on daily nurses, psychologists, social workers, and phys-
activities, sleep, mood, function, and quality of iotherapists may be needed in severe pain that is
life. not easily treated [42–44]. Furthermore, a close
It is a puzzle why some patients with DPN collaboration between diabetes treatment units
develop painful symptoms, while others do not and primary care physicians, which patients
[28, 29]. Most studies are cross-sectional, and attend regularly, and pain clinics may be helpful
there is generally a lack of longitudinal studies to and convenient for the patients.
elucidate the natural course of the disease and the
risk factors for developing neuropathic pain in
diabetes. Type 2 diabetes compared to type 1 dia- 3 Pharmacological Treatment
betes and higher measures of HbA1c have been
associated with P-DPN. Recent studies have also Currently there is no generally established dis-
found correlation of P-DPN with increased sever- ease modifying therapy for P-DPN and the aim of
ity of neuropathy [7, 8, 30–32]. Other risk factors the treatment is therefore mainly improvement of
that have been linked to P-DPN are smoking, modifiable risk factors, quality of life, and symp-
female sex, increasing age, obesity, and the co-­ tomatic pain relief.
occurrence of other diabetic complications [16,
33, 34]. The genetic component in the risk profile
of P-DPN is described in chapter “The Genomics 3.1 Pathogenesis Orientated
of Diabetic Neuropathy”. Treatment
Psychosocial factors such as reduced quality
of life, depression, sleep deprivation, and anxiety Optimal glycemic control and the assessment of
have consistently been shown to be associated vascular and lifestyle risk factors are important.
444 S. S. Gylfadottir and N. B. Finnerup

It is well known that hyperglycemia plays a role medication [59]. Additionally patients with
in the pathogeneses of neuropathy [45], and it P-DPN can be elderly and suffer from other
may contribute to the pathogeneses of neuro- chronic diseases or other macro- or microvascu-
pathic pain, although the evidence for improved lar diabetes complications, for example, heart
glycemic control as a pain relief is limited as and kidney diseases, potentially reinforcing the
there is a lack of randomized controlled studies side effects or rendering the recommended treat-
[43, 46, 47]. In a meta-analysis, enhanced glyce- ment contraindicated.
mic control was significantly effective in the pre- In 2016 the neuropathic pain special interest
vention of neuropathy in type 1 diabetes patients, group of the International Association for the
but not in type 2 diabetes [48]. A clinical study in Study of Pain (NeuPSIG) presented treatment
type 2 diabetes however showed improvement in guidelines for neuropathic pain based on system-
neurophysiological parameters with strict blood atic review and meta-analysis [59]. These recom-
sugar control without hypoglycemia for a longer mendations are based on a large number of
period of time in patients with poor glycemic studies and apply to patients with neuropathic
control [49]. pain of many etiologies, based on the conclusion
Alpha-lipoic acid (ALA) is suggested to be a made by the authors that the effect of the treat-
causative treatment of DPN by having beneficial ment does not depend on the etiology [59]. These
effects on vascular dysfunction, tissue hypoxia, recommendations are in line with other recom-
and glucose utilization. It is also suggested to mendations for painful DPN, from the European
prevent painful symptoms by possibly suppress- Federation of Neurological Societies (EFNS)
ing the activation of TRPV1 channel via activa- [60], the American Academy of Neurology [61],
tion of the NF-κB, a cellular nuclear transcription the Agency for Health Research and Quality
factor protein complex, but with limited effect (AHRQ) [62], the Toronto Expert Panel on
from clinical trials [50–53]. Another potential Diabetic Neuropathy [46], and a recent meta-­
neuroprotective agent in DPN is Actovegin that analysis on P-DPN [63].
consists of filtered extract from calf serum and is
thought to have antihypoxic effect on tissue. In a
multicenter, double blinded, randomized trial, 3.3 Guideline Recommendations
treatment with Actovegin improved symptoms of
neuropathy including pain, paresthesia, and Pregabalin and gabapentin, tricyclic antidepres-
numbness and the improvement was considered sants (TCA), and serotonin noradrenaline reup-
to be clinically relevant [54, 55]. take inhibitors (SNRIs) are recommended as a
Vitamin D has been thought to play a role in first-line therapy with a strong recommendation
peripheral neuropathy by stimulating keratino- and moderate to high quality of evidence [60,
cytes in the skin to produce nerve growth factor, 61, 63]. In terms of safety, there is no evidence
with D vitamin as a potential therapeutic agent to suggest one of these drugs is above others
[56]. No randomized, double-blinded studies [59, 64]. Duloxetine has been recommended for
have assessed the efficacy of vitamin D in painful DPN.  Capsaicin 8% and lidocaine 5%
P-DPN [57, 58]. patches are recommended as a second-line ther-
apy with moderate to high quality of evidence
and a week’s recommendation for use. Finally,
3.2 Symptomatic Treatment botulin toxin A subcutaneous injection is rec-
ommended as third-­ line treatment option
The challenges in the medical treatment of neu- (Table 1 and Fig. 3) [59].
ropathic pain lie in the relatively high numbers The results for combination therapy were
needed to treat (NNT) which is the number of inconclusive in both meta-analyses [59, 63],
patients needed to treat for one to have 50% pain although the search for sufficient pain relief in
reduction and often intolerable side effects of the the clinical setting often results in a combination
Characteristics and Treatment of Painful Diabetic Neuropathy 445

Table 1  Guidelines recommendation


Daily doses Common side effects Precautions
First-line recommendations
Tricyclic 25-(100) 150 mg, Sedation, dry mouth, constipation, People with seizures,
antidepressants starting with doses of blurred vision, weight gain, glaucoma, cardiac diseases,
(TCAs) 10–25 mg orthostatic hypotension with risk and dysuria and the elderly
of falls in elderly patients. >65 years.
ECG before starting
treatment for QT interval.
Serotonin-­ 60–120 mg for Nausea, somnolence, dizziness, In people with seizures,
norepinephrine duloxetine and constipation, dry mouth, glaucoma, uncontrolled
inhibitors (SNRIs) 150–225 mg for hypertension. Often mild and hypertension, and in the
venlafaxine in one transient side effects elderly.
dose
Pregabalin 300–600 mg in two Dizziness, somnolence, headache, Reduced doses in people
doses/day peripheral edema, and weight gain. with impaired kidney
Risk of suicidal thoughts function
Gabapentin 900–3600 mg in three Same as for pregabalin Same as for pregabalin
doses/day
Second-line recommendations
Lidocaine 5% One to three patches in Mild redness, swelling, irritation, Avoid contact with eyes,
patches the painful area up to change in skin color, and nose, and mouth
12 h numbness
Capsaicin 8% One to four patches in Local discomfort and skin Should not be used in the
patches the painful area for reaction/burning face and on or near mucous
30–60 min every membranes
3 months
Examples of SNRIs: Venlafaxine, Duloxetine, TCAs: Amitriptyline, Nortriptyline, Imipramine. For detailed informa-
tion on doses, side effects, and precautions, please refer to individual product information

of more than one of the recommended drugs, 3.4 Modes of Action,


preferable using drugs with different modes of Recommended Dose, and Side
action. The question whether it is better to Effects
increase the drug dose of a monotherapy with one
of the recommended drugs or if it is better to Tricyclic antidepressants (TCAs) act on the nor-
combine two drugs in patients that do not respond adrenergic system by inhibiting presynaptic
to a standard dose was addressed in the reuptake of monoamines (noradrenalin and sero-
COMBO-DN study of patients with P-DPN [65]. tonin) and the place of action for the TCAs is
The comparison between high recommended thought to be both central and peripheral [20, 67].
dose of pregabalin or duloxetine in patients that Other mechanisms may also be involved, includ-
did not respond to standard dose was compared ing blockade of voltage-gated sodium channels
to the effect of a combination therapy of the two and NMDA (N-methyl-d-aspartate) receptors
drugs at moderate dosages, with no differences [68, 69]. Experimental data have suggested that
between the groups, although the combination TCAs work through an effect on β2 adrenergic
therapy was considered well tolerated and safe receptors. However, a recent randomized con-
[65]. It is worth noting that patients with diabetes trolled trial found that the tricyclic antidepressant
often receive multiple medication for glycemic imipramine but not the β-agonist terbutaline had
control, hypertension, and dyslipidemia and in effect on painful polyneuropathy suggesting that
some cases medication for sleep disturbances, β2 agonisms are not a clinical important mecha-
depression, or anxiety. Polypharmacy may there- nism of action of TCAs in painful polyneuropa-
fore raise concerns about compliance, adherence, thy [70]. Examples of TCAs are amitriptyline,
and drug–drug interactions [66]. nortriptyline, and imipramine and the overall
446 S. S. Gylfadottir and N. B. Finnerup

Painful diabetic polyneuropathy

Before initiating drug treatment asess


comorbidity, contraindictations and side effects.

First line drug therapy

TCA Non-pharmacological treatment


SNRIs Provide patient education, encourage self-care
and consider psychological help and
Gabapentin/pregabalin physiotherapy.

If there is partiel, but inadequate pain


control with monotherapy with one ore
more of the first line therapy options in a
recommended dose, the combination of
TCA+gabapentin or pregabalin, or
SNRIs+gabapentin or pregabalin can be
considered.

Second line drug therapy

Lidocain 5% patches

Capsacain 8% patches
Topical treatment can be indicated in a
selected group of patients.

Fig. 3  Treatment algorithm for P-DPN

number of patients needed to treat (NNT) to tors than amitriptyline and may therefore cause
achieve at least 50% reduction in pain intensity less sedative side effects [69, 71]. Caution should
for one patient for TCAs in neuropathic pain was be shown in the treatment of elderly people
3.6 (95%CI 3.0–4.4) where most trials included >65 years where lower doses are required because
amitriptyline [59]. A starting dose of 10–25 mg of risk of sedation, falls, and sudden cardiac
increasing with 10  mg every fifth day until death [72, 73].
desired pain relief is obtained where maximum SNRIs also target the serotonin noradrenergic
dose of 100–150 mg is recommended. The most system by inhibiting presynaptic reuptake of sero-
common side effects are dry mouth, constipation, tonin and noradrenalin, augmenting descending
sedation, blurred vision, weight gain, and ortho- inhibitory pathways. Examples are duloxetine and
static hypotension with the risk of falls in elderly venlafaxine. Most common adverse effects are
patients. An ECG before treatment is recom- nausea, somnolence, dizziness, constipation, and
mended to exclude prolongation of PR and QT dry mouth; these side effects are often mild and
intervals. Secondary amines (nortriptyline and transient. Recommended daily doses are
desipramine) have lower affinity for muscarinic 60–120 mg/day for duloxetine and 150–225 mg/
acetylcholine receptors and imipramine has day for venlafaxine. The NNT for 50% pain reduc-
lower affinity to block histaminergic H1 recep- tion for SNRIs is 6.4 (95% CI 5.2–8.4) [20, 59].
Characteristics and Treatment of Painful Diabetic Neuropathy 447

The group of antidepressants should be tried [76, 78]. Tramadol, which is a centrally acting
for some time for pain relief before switching to synthetic opioid working on both opioid and
another drug because lack of effect [69]. In monoaminergic pathways, may be considered
patients with concomitant depression TCAs and for flares of pain. A range of drugs have incon-
SNRIs may have some effect on depressive clusive evidence based on conflicting results
symptoms [41]. from randomized trials, including lamotrigine,
Gabapentin and pregabalin have similar oxcarbazepine, topiramate, and selective sero-
modes of action and bind to the α2δ subunit of tonin-reuptake inhibitors, and thus there is no
the presynaptic voltage-gated calcium channels evidence to support their use for P-DPN, and
and thereby reduce Ca2+ influx resulting in further studies are needed to support their use,
decreased neurotransmitter release [20, 67]. The e.g. in specific subtypes of patients [59]. One
site of action can be peripheral, spinal, and study found oxcarbamazepine to be effective in
supraspinal. The NNT for pregabalin is 7.7 (6.5; a subgroup of patients with painful polyneu-
9.4) and for gabapentin 7.2 (5.9; 9.1) [59]. ropathy and a sensory phenotype with pre-
Common side effects are dizziness, somnolence, served small-fiber sensation (cold and warm
peripheral edema, headache, and weight gain. sensation and sensory hypersensitivity com-
Daily doses for gabapentin are 900–3600  mg pared to those without), but these results have
daily in three doses starting with 300  mg/day not been confirmed [79]. Recent systematic
and for pregabalin 300–600  mg in two doses reviews find no evidence for the use of cannabi-
starting with 75  mg/day. There are increasing noids, cannabis, and cannabis-based medicines
concerns and evidence for misuse of gabapentin in chronic pain [80, 81]. The NeuPSIG guide-
and pregabalin [74]. lines have a weak recommendation against the
Topical treatment with capsaicin 8% and lido- use of cannabis-based medicines in neuropathic
caine 5% patches can be beneficial in a selected pain because of lack of effect in clinical trials,
group of patients. Capsaicin binds to the transient potential misuse, diversion, and long-term
receptor potential vanilloid 1 (TRPV1) on noci- mental health risks [59].
ceptors desensitizing them for 12  weeks.
Normally one to four patches are applied in the
painful area for 30–60  min every 3  months. 3.6 Emerging Treatment
Lidocaine inhibits ectopic nerve activity by Possibilities
blocking voltage-gated sodium channels [20].
One to three lidocaine 5% patches are applied in Several drug classes are currently in clinical
the painful area for up to 12 h. development for neuropathic pain, although to
date, we have seen a failure in developing new
drug classes for neuropathic pain [82, 83]. A
3.5 Other Pharmacological peripherally acting drug EMA401, which is a
Treatment Options competitive antagonist to angiotensin II type 2
receptor, showed promising effects in peripheral
Tramadol and strong opioids have also shown neuropathic pain [84], but recently phase 2 trials
some effect in randomized controlled trials of have been terminated due to safety reasons.
short duration, but are generally not recom- Antibodies targeting the activity of human nerve
mended for the treatment of chronic non-­ growth factors are also in development for pain.
malignant pain because of long-term Recombinant human nerve growth factor had no
consequences, risk of abuse and dependence, effect on diabetic polyneuropathy in a large trial
and lack of effect on functional status [75–78]. but showed some effect on pain in the legs, which
If opioids are indicated for medium-term use, a was one of many secondary outcomes [85].
low dose in a monitored setting following local Tanezumab, a humanized monoclonal antibody
opioid prescribing guidelines is recommended against nerve growth factor was found to reduce
448 S. S. Gylfadottir and N. B. Finnerup

P-DPN despite early study termination due to a may have a place in the treatment of painful DPN
partial clinical hold by the US Food and Drug although the evidence is sparse.
Administration (FDA) due to joint-related safety Physiotherapy treatment options can be aimed
[86], but currently the drug is only being devel- at pain reduction, preserve function level, and
oped for osteoarthritis and other pain types. prevent immobilization or it can consist of thera-
Subtype-selective sodium channel inhibitors, in peutic exercise [91]. In a qualitative study,
particular Nav1.7 blockers, are also being investi- patients with P-DPN described walking difficul-
gated for neuropathic pain. ties and other impacts of pain on physical func-
tions [92]. A review of physical therapy in
chemotherapy induced neuropathy showed that
4 Nonpharmacological there was possible a symptom effect, although
Treatment with limited evidence [93].
In general, physical activity in adjutancy to
The first treatment of neuropathic pain offered to other treatments or alone could have positive
patients is most often pharmacological, but it is effects on P-DPN, although the evidence is lim-
also important to consider nonpharmacological ited with a high risk of bias [94, 95].
treatment options, either as the only treatment or Yoga, mindfulness, and meditation are gain-
as an addition to medical treatment. ing increased popularity as a treatment option in
With a general agreement of having P-DPN various conditions, often for the improvement of
being associated with worse mental health, there mental health and quality of life, but data on the
are paradoxically few studies that have examined efficacy for pain relief are few or missing in the
the effect of psychological treatment in P-DPN treatment of neuropathic pain, the same is for
[39, 87]. Psychological therapy for chronic pain acupuncture [95–97].
may consist of cognitive behavioral therapy Neurostimulation has been used as a treatment
(CBT), a traditional therapy form that helps option in P-DPN. Examples are spinal cord stim-
patients to identify, change, and provide coping ulation (SCS) and transcutaneous electrical stim-
strategies for negative thoughts or behavior asso- ulation (TENS). According to the newly
ciated with pain. Another therapy form derived published guidelines from the European Academy
from CBT also used in the treatment of pain is of Neurology (EAN), there is weak evidence for
acceptance and commitment therapy (ACT) adding spinal cord stimulation (SCS) to the phar-
which integrates acceptance, cognitive defusion, macological treatment in P-DPN compared with
and mindfulness to produce more psychological pharmacological treatment alone [98] and a
flexibility [88]. In a recently published single arm recent meta-analysis including randomized con-
primarily feasibility trial with online ACT treat- trolled trials concluded that SCS was effective for
ment in patients with P-DPN the clinical out- pain relief compared to standard care [99]. A
comes showed beneficial effect on psychological Cochrane review of TENS treatment for neuro-
functions and pain intensity, although the study pathic pain could not conclude whether the treat-
has limitations due to design [89]. In a newly ment was effective compared to sham therapy or
updated systematic review of the effect of psy- not, because of the size and quality of the included
chological therapies on the broad spectrum of studies, neither were side effects nor adverse
chronic pain, compared to standard treatment or events adequately addressed in the studies [100].
waiting list, there was only very small to small
beneficial effect of CBT in pain reduction, dis-
ability, and stress [90]. In separate studies of neu- 5 Conclusion
ropathic pain and P-DPN the evidence was too
sparse and insufficient to make any conclusions Painful neuropathy is a common and often dis-
on the effect of psychological treatment in pain abling consequence of diabetes. It is a neuro-
relief [39, 87]. Therefore psychological treatment pathic pain and is caused by the lesion of the
Characteristics and Treatment of Painful Diabetic Neuropathy 449

peripheral nerves. The pharmacological treat- ments for diabetic polyneuropathy. Endocr Rev.
2019;40:153–92.
ment is challenging and often provides only par- 14. Partanen J, Niskanen L, Lehtinen J, Mervaala
tial pain relief at best. Therefore both E, Siitonen O, Uusitupa M.  Natural history of
nonpharmacological and pharmacological treat- peripheral neuropathy in patients with non-insu-
ments are recommended. lin-dependent diabetes mellitus. N Engl J Med.
1995;333:89–94.
15. Ziegler D, Strom A, Lobmann R, Reiners K, Rett
K, Schnell O.  High prevalence of diagnosed and
References undiagnosed polyneuropathy in subjects with and
without diabetes participating in a nationwide edu-
1. Izenberg A, Perkins BA, Bril V.  Diabetic neuropa- cational initiative (PROTECT study). J Diabetes
thies. Semin Neurol. 2015;35:424–30. Complicat. 2015;29:998–1002.
2. Gylfadottir SS, Weeracharoenkul D, Andersen 16. Truini A, Spallone V, Morganti R, et al. A cross sec-
ST, Niruthisard S, Suwanwalaikorn S, Jensen tional study investigating frequency and features of
TS.  Painful and non-painful diabetic polyneuropa- definitely diagnosed diabetic painful polyneuropa-
thy: clinical characteristics and diagnostic issues. J thy. Pain. 2018;159:2658.
Diabetes Investig. 2019;10:1148–57. 17. Van Acker K, Bouhassira D, De Bacquer D, et  al.
3. Raja SN, Carr DB, Cohen M, et  al. The revised Prevalence and impact on quality of life of periph-
International Association for the Study of Pain defi- eral neuropathy with or without neuropathic
nition of pain: concepts, challenges, and compro- pain in type 1 and type 2 diabetic patients attend-
mises. Pain. 2020;161:1976. ing hospital outpatients clinics. Diabetes Metab.
4. Taxonomy I. https://www.iasp-­pain.org/terminology 2009;35:206–13.
?navItemNumber=576 18. Aslam A, Singh J, Rajbhandari S.  Prevalence of
5. Tesfaye S, Boulton AJ, Dyck PJ, et al. Diabetic neu- painful diabetic neuropathy using the self-completed
ropathies: update on definitions, diagnostic criteria, Leeds assessment of neuropathic symptoms and
estimation of severity, and treatments. Diabetes signs questionnaire in a population with diabetes.
Care. 2010;33:2285–93. Can J Diabetes. 2015;39:285–95.
6. Callaghan BC, Cheng HT, Stables CL, Smith AL, 19. Bouhassira D, Letanoux M, Hartemann A. Chronic
Feldman EL.  Diabetic neuropathy: clinical mani- pain with neuropathic characteristics in diabetic
festations and current treatments. Lancet Neurol. patients: a French cross-sectional study. PLoS One.
2012;11:521–34. 2013;8:e74195.
7. Themistocleous AC, Ramirez JD, Shillo PR, et  al. 20. Finnerup NB, Kuner R, Jensen TS.  Neuropathic
The Pain in Neuropathy Study (PiNS): a cross-­ pain: from mechanisms to treatment. Physiol Rev.
sectional observational study determining the 2020;101:259.
somatosensory phenotype of painful and painless 21. Finnerup NB, Haroutounian S, Kamerman P, et  al.
diabetic neuropathy. Pain. 2016;157:1132–45. Neuropathic pain: an updated grading system for
8. Abbott CA, Malik RA, van Ross ER, Kulkarni J, research and clinical practice. Pain. 2016;157:1599.
Boulton AJ. Prevalence and characteristics of pain- 22. Treede RD, Jensen TS, Campbell JN, et  al.
ful diabetic neuropathy in a large community-based Neuropathic pain: redefinition and a grading sys-
diabetic population in the U.K.  Diabetes Care. tem for clinical and research purposes. Neurology.
2011;34:2220–4. 2008;70:1630–5.
9. Andersen ST, Witte DR, Dalsgaard EM, et al. Risk 23. Attal N, Bouhassira D, Baron R.  Diagnosis and
factors for incident diabetic polyneuropathy in a assessment of neuropathic pain through question-
cohort with screen-detected type 2 diabetes followed naires. Lancet Neurol. 2018;17:456–66.
for 13 years: addition-Denmark. Diabetes Care. 24. Bouhassira D, Attal N, Alchaar H, et al. Comparison
2018;41:1068–75. of pain syndromes associated with nervous or
10. Gylfadottir SS, et  al. Diagnosis and prevalence of somatic lesions and development of a new neuro-
diabetic polyneuropathy: a cross-sectional study of pathic pain diagnostic questionnaire (DN4). Pain.
Danish patients with type 2 diabetes. Eur J Neurol. 2005;114:29–36.
2020;27:2575. 25. Spallone V, Morganti R, D’Amato C, Greco C,
11. van Hecke O, Austin SK, Khan RA, Smith BH, Cacciotti L, Marfia GA.  Validation of DN4 as a
Torrance N. Neuropathic pain in the general popula- screening tool for neuropathic pain in painful dia-
tion: a systematic review of epidemiological studies. betic polyneuropathy. Diabet Med. 2012;29:578–85.
Pain. 2014;155:654–62. 26. Bouhassira D, Attal N, Fermanian J, et  al.
12. Feldman EL, Callaghan BC, Pop-Busui R, et  al. Development and validation of the neuropathic pain
Diabetic neuropathy. Nat Rev Dis Primers. symptom inventory. Pain. 2004;108:248–57.
2019;5:41. 27. Jensen MP, Karoly P, Braver S. The measurement of
13. Bonhof GJ, Herder C, Strom A, Papanas N, Roden clinical pain intensity: a comparison of six methods.
M, Ziegler D. Emerging biomarkers, tools, and treat- Pain. 1986;27:117–26.
450 S. S. Gylfadottir and N. B. Finnerup

28. Spallone V, Greco C.  Painful and painless diabetic 43. Sloan G, Shillo P, Selvarajah D, et  al. A new look
neuropathy: one disease or two? Curr Diab Rep. at painful diabetic neuropathy. Diabetes Res Clin
2013;13:533–49. Pract. 2018;144:177–91.
29. Shillo P, Sloan G, Greig M, et al. Painful and pain- 44. Shaygan M, Böger A, Kröner-Herwig B. Predicting
less diabetic neuropathies: what is the difference? factors of outcome in multidisciplinary treat-
Curr Diab Rep. 2019;19:32. ment of chronic neuropathic pain. J Pain Res.
30. Tesfaye S, Boulton AJ, Dickenson AH. Mechanisms 2018;11:2433–43.
and management of diabetic painful distal 45. Calcutt NA.  Diabetic neuropathy and neuropathic
symmetrical polyneuropathy. Diabetes Care. pain: a (con)fusion of pathogenic mechanisms? Pain.
2013;36:2456–65. 2020;161:S65–s86.
31. Davies M, Brophy S, Williams R, Taylor A.  The 46. Tesfaye S, Vileikyte L, Rayman G, et al. Painful dia-
prevalence, severity, and impact of painful diabetic betic peripheral neuropathy: consensus recommen-
peripheral neuropathy in type 2 diabetes. Diabetes dations on diagnosis, assessment and management.
Care. 2006;29:1518–22. Diabetes Metab Res Rev. 2011;27:629–38.
32. Raputova J, Srotova I, Vlckova E, et  al. Sensory 47. Boulton AJ, Drury J, Clarke B, Ward JD. Continuous
phenotype and risk factors for painful diabetic neu- subcutaneous insulin infusion in the management
ropathy: a cross-sectional observational study. Pain. of painful diabetic neuropathy. Diabetes Care.
2017;158:2340–53. 1982;5:386–90.
33. Gylfadottir SS, Christensen DH, Nicolaisen SK, 48. Callaghan BC, Little AA, Feldman EL, Hughes
et al. Diabetic polyneuropathy and pain, prevalence, RA.  Enhanced glucose control for preventing and
and patient characteristics: a cross-sectional ques- treating diabetic neuropathy. Cochrane Database
tionnaire study of 5,514 patients with recently diag- Syst Rev. 2012;6:CD007543.
nosed type 2 diabetes. Pain. 2019;161:574. 49. Ishibashi F, Taniguchi M, Kosaka A, Uetake H,
34. Hebert HL, Veluchamy A, Torrance N, Smith Tavakoli M.  Improvement in neuropathy outcomes
BH.  Risk factors for neuropathic pain in diabetes with normalizing HbA(1c) in patients with type 2
mellitus. Pain. 2016;158:560. diabetes. Diabetes Care. 2019;42:110–8.
35. Naranjo C, Del Reguero L, Moratalla G, Hercberg 50. Papanas N, Ziegler D. Emerging drugs for diabetic
M, Valenzuela M, Failde I. Anxiety, depression and peripheral neuropathy and neuropathic pain. Expert
sleep disorders in patients with diabetic neuropathic Opin Emerg Drugs. 2016;21:393–407.
pain: a systematic review. Expert Rev Neurother. 51. Papanas N, Ziegler D. Efficacy of alpha-lipoic acid
2019;19:1201–9. in diabetic neuropathy. Expert Opin Pharmacother.
36. Gore M, Brandenburg NA, Dukes E, Hoffman DL, 2014;15:2721–31.
Tai KS, Stacey B.  Pain severity in diabetic periph- 52. Zhang BY, Zhang YL, Sun Q, et  al. Alpha-lipoic
eral neuropathy is associated with patient function- acid downregulates TRPV1 receptor via NF-κB and
ing, symptom levels of anxiety and depression, and attenuates neuropathic pain in rats with diabetes.
sleep. J Pain Symptom Manag. 2005;30:374–85. CNS Neurosci Ther. 2020;26:762–72.
37. D’Amato C, Morganti R, Greco C, et  al. Diabetic 53. Ziegler D, Nowak H, Kempler P, Vargha P, Low
peripheral neuropathic pain is a stronger predictor PA. Treatment of symptomatic diabetic polyneurop-
of depression than other diabetic complications and athy with the antioxidant alpha-lipoic acid: a meta-­
comorbidities. Diab Vasc Dis Res. 2016;13:418–28. analysis. Diabet Med. 2004;21:114–21.
38. Gylfadottir SS, Christensen DH, Nicolaisen SK, 54. Ziegler D, Edmundson S, Gurieva I, Mankovsky B,
et al. Diabetic polyneuropathy and pain, prevalence, Papanas N, Strokov I. Predictors of response to treat-
and patient characteristics: a cross-sectional ques- ment with actovegin for 6 months in patients with
tionnaire study of 5,514 patients with recently diag- type 2 diabetes and symptomatic polyneuropathy. J
nosed type 2 diabetes. Pain. 2020;161:574–83. Diabetes Complicat. 2017;31:1181–7.
39. Kioskli K, Scott W, Winkley K, Kylakos S, 55. Ziegler D, Movsesyan L, Mankovsky B, Gurieva I,
McCracken LM.  Psychosocial factors in pain- Abylaiuly Z, Strokov I.  Treatment of symptomatic
ful diabetic neuropathy: a systematic review of polyneuropathy with actovegin in type 2 diabetic
treatment trials and survey studies. Pain Med. patients. Diabetes Care. 2009;32:1479–84.
2019;20:1756–73. 56. Fukuoka M, Sakurai K, Ohta T, Kiyoki M, Katayama
40. Geelen CC, Smeets R, Schmitz S, van den Bergh JP, I.  Tacalcitol, an active vitamin D3, induces nerve
Goossens M, Verbunt JA. Anxiety affects disability growth factor production in human epidermal
and quality of life in patients with painful diabetic keratinocytes. Skin Pharmacol Appl Ski Physiol.
neuropathy. Eur J Pain. 2017;21:1632–41. 2001;14:226–33.
41. Finnerup NB. Nonnarcotic methods of pain manage- 57. Shillo P, Selvarajah D, Greig M, et al. Reduced vita-
ment. N Engl J Med. 2019;380:2440–8. min D levels in painful diabetic peripheral neuropa-
42. Scascighini L, Toma V, Dober-Spielmann S, Sprott thy. Diabet Med. 2019;36:44–51.
H.  Multidisciplinary treatment for chronic pain: a 58. Yammine K, Wehbe R, Assi C.  A systematic
systematic review of interventions and outcomes. review on the efficacy of vitamin D supplementa-
Rheumatology (Oxford). 2008;47:670–8.
Characteristics and Treatment of Painful Diabetic Neuropathy 451

tion on diabetic peripheral neuropathy. Clin Nutr. of pregabalin and gabapentin: a systematic review
2020;39:2970–4. update. Drugs. 2020;81:125.
59. Finnerup NB, Attal N, Haroutounian S, et  al. 75. Franklin GM.  Opioids for chronic noncancer pain:
Pharmacotherapy for neuropathic pain in adults: a a position paper of the American Academy of
systematic review and meta-analysis. Lancet Neurol. Neurology. Neurology. 2014;83:1277–84.
2015;14:162–73. 76. Dowell D, Haegerich TM, Chou R. CDC guideline
60. Attal N, Cruccu G, Baron R, et al. EFNS guidelines on for prescribing opioids for chronic pain--United
the pharmacological treatment of neuropathic pain: States, 2016. JAMA. 2016;315:1624–45.
2010 revision. Eur J Neurol. 2010;17:1113–e1188. 77. Hoffman EM, Watson JC, St Sauver J, Staff NP,
61. Bril V, England J, Franklin GM, et al. Evidence-based Klein CJ. Association of Long-term Opioid Therapy
guideline: treatment of painful diabetic neuropa- With Functional Status, adverse outcomes, and mor-
thy: report of the American Academy of Neurology, tality among patients with polyneuropathy. JAMA
the American Association of Neuromuscular and Neurol. 2017;74:773–9.
Electrodiagnostic Medicine, and the American 78. Ballantyne JCBS, Blyth F, Cordosa M, Finley
Academy of Physical Medicine and Rehabilitation. A, Furlan A, et  al. https://www.iasp-­pain.org/
Neurology. 2011;76:1758–65. Advocacy/OpioidPositionStatement.2018. Accessed
62. Agency for Health Research and Quality (AHRQ). 19 Jan 2021.
Accessed 15 Jan 2021. 79. Demant DT, Lund K, Vollert J, et  al. The effect
63. Waldfogel JM, Nesbit SA, Dy SM, et  al. of oxcarbazepine in peripheral neuropathic pain
Pharmacotherapy for diabetic peripheral neuropa- depends on pain phenotype: a randomised, double-­
thy pain and quality of life: a systematic review. blind, placebo-controlled phenotype-stratified study.
Neurology. 2017;88:1958–67. Pain. 2014;155:2263–73.
64. Enomoto H, Yasuda H, Nishiyori A, et al. Duloxetine 80. Fisher E, Moore RA, Fogarty AE, et al. Cannabinoids,
in patients with diabetic peripheral neuropathic pain cannabis, and cannabis-based medicine for pain
in Japan: a randomized, doubleblind, noninferior- management: a systematic review of randomised
ity comparative study with pregabalin. J Pain Res. controlled trials. Pain. 2020;162:S45.
2018;11:1857–68. 81. Moore RA, Fisher E, Finn DP, et al. Cannabinoids,
65. Tesfaye S, Wilhelm S, Lledo A, et al. Duloxetine and cannabis, and cannabis-based medicines for pain
pregabalin: high-dose monotherapy or their combi- management: an overview of systematic reviews.
nation? The “COMBO-DN study”--a multinational, Pain. 2020;162:S67.
randomized, double-blind, parallel-group study in 82. Attal N, Bouhassira D.  Translational neuropathic
patients with diabetic peripheral neuropathic pain. pain research. Pain. 2019;160(Suppl 1):S23–s28.
Pain. 2013;154:2616–25. 83. Yekkirala AS, Roberson DP, Bean BP, Woolf
66. Finnerup NB. Add-on therapy: when two are not bet- CJ. Breaking barriers to novel analgesic drug devel-
ter than one. Pain. 2013;154:2579–80. opment. Nat Rev Drug Discov. 2017;16:545–64.
67. Kremer M, Salvat E, Muller A, Yalcin I, Barrot 84. Rice ASC, Dworkin RH, McCarthy TD, et  al.
M.  Antidepressants and gabapentinoids in neuro- EMA401, an orally administered highly selective
pathic pain: mechanistic insights. Neuroscience. angiotensin II type 2 receptor antagonist, as a novel
2016;338:183–206. treatment for postherpetic neuralgia: a randomised,
68. Javed S, Alam U, Malik RA.  Burning through the double-blind, placebo-controlled phase 2 clinical
pain: treatments for diabetic neuropathy. Diabetes trial. Lancet. 2014;383:1637–47.
Obes Metab. 2015;17:1115–25. 85. Apfel SC, Schwartz S, Adornato BT, et al. Efficacy
69. Sindrup SH, Otto M, Finnerup NB, Jensen and safety of recombinant human nerve growth factor
TS.  Antidepressants in the treatment of neu- in patients with diabetic polyneuropathy: a random-
ropathic pain. Basic Clin Pharmacol Toxicol. ized controlled trial. rhNGF Clinical Investigator
2005;96:399–409. Group. JAMA. 2000;284:2215–21.
70. Gillving M, Demant D, Holbech JV, et al. A random- 86. Bramson C, Herrmann DN, Carey W, et  al.
ized, controlled trial of a β2-agonist in painful poly- Exploring the role of tanezumab as a novel treat-
neuropathy. Pain. 2020;162:1364. ment for the relief of neuropathic pain. Pain Med.
71. Gillman PK.  Tricyclic antidepressant pharmacol- 2015;16:1163–76.
ogy and therapeutic drug interactions updated. Br J 87. Eccleston C, Hearn L, Williams AC. Psychological
Pharmacol. 2007;151:737–48. therapies for the management of chronic neuro-
72. American Geriatrics Society. Updated beers criteria pathic pain in adults. Cochrane Database Syst Rev.
for potentially inappropriate medication use in older 2015;2015:CD011259.
adults. J Am Geriatr Soc. 2015;2015(63):2227–46. 88. Hayes SC, Luoma JB, Bond FW, Masuda A, Lillis
73. Ray WA, Meredith S, Thapa PB, Hall K, Murray J. Acceptance and commitment therapy: model, pro-
KT. Cyclic antidepressants and the risk of sudden car- cesses and outcomes. Behav Res Ther. 2006;44:1–25.
diac death. Clin Pharmacol Ther. 2004;75:234–41. 89. Kioskli K, Scott W, Winkley K, Godfrey E,
74. Evoy KE, Sadrameli S, Contreras J, Covvey JR, McCracken LM. Online acceptance and commitment
Peckham AM, Morrison MD.  Abuse and misuse therapy for people with painful diabetic neuropathy
452 S. S. Gylfadottir and N. B. Finnerup

in the United Kingdom: a single-arm feasibility trial. painful diabetic neuropathy--a systematic review of
Pain Med. 2020;21:2777–88. the literature. Physiotherapy. 2015;101:319–26.
90. Williams ACC, Fisher E, Hearn L, Eccleston 96. Baute V, Zelnik D, Curtis J, Sadeghifar
C.  Psychological therapies for the management F.  Complementary and alternative medicine for
of chronic pain (excluding headache) in adults. painful peripheral neuropathy. Curr Treat Options
Cochrane Database Syst Rev. 2020;8:CD007407. Neurol. 2019;21:44.
91. Akyuz G, Kenis O.  Physical therapy modalities 97. van Laake-Geelen CCM, Smeets R, Quadflieg S,
and rehabilitation techniques in the management Kleijnen J, Verbunt JA. The effect of exercise ther-
of neuropathic pain. Am J Phys Med Rehabil. apy combined with psychological therapy on physi-
2014;93:253–9. cal activity and quality of life in patients with painful
92. Brod M, Pohlman B, Blum SI, Ramasamy A, diabetic neuropathy: a systematic review. Scand J
Carson R.  Burden of illness of diabetic periph- Pain. 2019;19:433–9.
eral neuropathic pain: a qualitative study. Patient. 98. Cruccu G, Garcia-Larrea L, Hansson P, et al. EAN
2015;8:339–48. guidelines on central neurostimulation therapy in
93. Jesson T, Runge N, Schmid AB. Physiotherapy for chronic pain conditions. Eur J Neurol. 2016;23:1489.
people with painful peripheral neuropathies: a nar- 99. Amato Nesbit S, Sharma R, Waldfogel JM, et  al.
rative review of its efficacy and safety. Pain Rep. Non-pharmacologic treatments for symptoms of
2020;5:1–e834. diabetic peripheral neuropathy: a systematic review.
94. Singleton JR, Smith AG, Marcus RL.  Exercise as Curr Med Res Opin. 2019;35:15–25.
therapy for diabetic and prediabetic neuropathy. 100. Gibson W, Wand BM, O’Connell NE.
Curr Diab Rep. 2015;15:120. Transcutaneous electrical nerve stimulation (TENS)
95. Davies B, Cramp F, Gauntlett-Gilbert J, Wynick D, for neuropathic pain in adults. Cochrane Database
McCabe CS. The role of physical activity and psy- Syst Rev. 2017;9:CD011976.
chological coping strategies in the management of
Orthostatic Hypotension
and Sudomotor Dysfunction
in Diabetes

Lauren F. Fanty and Christopher H. Gibbons

1 Introduction mates ranging from 7% to 90% [10]. Prevalence


of autonomic impairment also varies between the
Diabetic autonomic neuropathy (DAN) is the types of diabetes, with an estimated prevalence of
leading cause of autonomic neuropathy in the 54% and 73% in type 1 and type 2 diabetes,
USA. There are multiple mechanisms underlying respectively [10]. Cardiac autonomic neuropathy
the pathophysiology of diabetic neuropathy (CAN) is the most life-threatening form of dia-
including neurovascular insufficiency, metabolic betic autonomic neuropathy with a 56% 10-year
insult to nerve fibers, neurohormonal growth fac- mortality [5, 10, 11]. In CAN, Vagus nerve dener-
tor insufficiency, and bioenergy failure [1]. vation leads to reduced heart rate variability and
Generalized DAN typically is gradual in onset presents initially as a resting tachycardia [6].
and leads to progressive autonomic impairment With continued disease progression, sympathetic
to vasomotor (vascular), visceromotor (gastroin- cardiac denervation leads to orthostatic hypoten-
testinal), parasympathetic, sympathetic adrener- sion and chronotropic incompetence [6, 12, 13].
gic, and sympathetic cholinergic autonomic Sudomotor (sweating) dysfunction is often
fibers [2–4]. Clinical features are broad, as DAN comorbid with CAN and one of the earliest signs
affects the cardiovascular, gastrointestinal, pupil- of DAN. Sudomotor dysfunction typically occurs
lomotor, thermoregulatory, sudomotor, and uro- in a length-dependent fashion, often in parallel
genital systems. Many large epidemiologic with sensory neuropathy.
studies have established the increased morbidity, In DAN, orthostatic hypotension is due to
mortality, and sudden death in patients with dia- sympathetic adrenergic failure, while sudomotor
betic autonomic neuropathy [4–9]. dysfunction function is due to sympathetic cho-
The prevalence of diabetic autonomic neurop- linergic failure [6]. This chapter will focus on the
athy varies depending on diagnostic criteria, pathophysiology, clinical presentation, diagnosis,
patient cohort, and testing modality with esti- and treatment of orthostatic hypotension and
sudomotor dysfunction in patients with diabetes.
L. F. Fanty
Department of Neurology, Beth Israel Deaconess
Medical Center, Harvard Medical School, 2 Orthostatic Hypotension
Boston, MA, USA
C. H. Gibbons (*) Orthostatic hypotension, OH, is defined as a fall
Beth Israel Deaconess Medical Center, in systolic blood pressure (SBP) of 20 mmHg or
Boston, MA, USA fall in diastolic blood pressure (DBP) of 10 mmHg
e-mail: cgibbons@bidmc.harvard.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 453
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_26
454 L. F. Fanty and C. H. Gibbons

within 3  min of standing or a head-up tilt to at known as postprandial hypotension. Postprandial


least 60° on a tilt table [14]. It is one of the most hypotension is defined as a fall of at least 10 mmHg
incapacitating manifestations of autonomic fail- within 2 h of eating [22]. This is caused by degen-
ure [6]. Orthostatic hypotension in isolation may eration of the fibers supplying the splanchnic mes-
be a clinical sign indicating a variety of medical enteric bed, a large vascular bed that increases in
conditions such as severe anemia, intravascular volume by 200–300% after a carbohydrate heavy
volume depletion, physical deconditioning, anti- meal, resulting in orthostatic stress from venous
hypertensive medication use, or impairment of pooling [22]. Normally, this increase in volume is
peripheral vasoconstriction [15, 16]. compensated for by an increase in sympathetic
Neurogenic orthostatic hypotension, nOH, is a response leading to splanchnic vasoconstriction
subset of OH that occurs when the autonomic ner- [19]. However, this compensatory mechanism is
vous system fails to provide sufficient autonomic deficient in patients with nOH.
postural response via sympathetic vasoconstric- Supine hypertension is another common com-
tion and fails to increase heart rate to maintain plication detected in an estimated 50% of patients
blood pressure [15]. In diabetes, there is degener- with nOH [15]. Supine hypertension is defined as
ation of sympathetic pre- and postganglionic a supine SBP  ≧  150 or DBP  ≧  90 [15, 16]. In
fibers leading to failure in both the vagal and patients with supine hypertension, a reduction in
adrenergic limbs of the baroreflex resulting in SBP of 30 mmHg or DBP of 15 mmHg may be a
nOH [6]. In a normal response to moving to the more appropriate criterion for nOH [15]. Supine
upright position, there is an increase in plasma hypertension is a product of both impaired baro-
norepinephrine as well as increased cardiac out- reflex function and chronically increased activa-
put and peripheral vasoconstriction. In nOH, there tion of the renin–angiotensin system from
is impaired release of peripheral norepinephrine orthostatic hypotension [15, 19]. nOH and supine
with impaired peripheral vasoconstriction and hypertension are associated conditions with the
impaired cardiac acceleration in response to a fall same underlying pathophysiology but create a
in blood pressure [17, 18]. Inadequate blood pres- challenge to effectively treat both conditions
sure response is most often attributable to simultaneously [15, 19].
decreased norepinephrine released from postgan- While our focus of discussion will be on nOH
glionic sympathetic nerves leading to pooling of secondary to disruption of peripheral sympatheti-
blood in the splanchnic region, pelvis, and depen- cally mediated reflex vasoconstriction attributed
dent areas [15]. Compensatory increase in heart to diabetic neuropathy, it is important to note that
rate when standing from a seated position is aug- other neuropathies, neurodegenerative disorders,
mented by norepinephrine and sympathetic nerve and high spinal cord lesions also affect peripheral
endings, which may also be impaired with sympa- sympathetic neurons [19]. Some neurodegenera-
thetic dysfunction [6, 19]. The Rochester Diabetic tive disorders, such as multiple systems atrophy,
Study, which evaluated a cohort of 83 Type I dia- affect central sympathetic neurons resulting in
betics and 148 Type II diabetics, found that of the nOH from central autonomic dysfunction, which
approximately 50% of patient with clinical mani- will not be a focus of this chapter [15].
festations of neuropathy, about 10% additionally
had clinical evidence of autonomic neuropathy
[20, 21]. Orthostatic hypotension was present in 3 Clinical Presentation
8.4% of type 1 diabetics and 7.4% of type 2 dia- and Initial Evaluation
betics [2, 20, 21]. Other estimates report the prev-
alence of orthostatic hypotension to be as high as The prevalence of orthostatic hypotension in dia-
20–25% in patients with long term diabetes [2]. betes is estimated to range from 6% to 30% and
A symptom that is sometimes associated with increases with age, disease duration, and disease
nOH is worsening of orthostatic tolerance after control [2, 6, 23]. It is more common in institution-
eating secondary to a drop in blood pressure, alized than community dwelling elderly and is
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 455

normally a late manifestation of cardiovascular the upper body, fatigue, dyspnea, chest pain, and
autonomic neuropathy [10]. Neurogenic OH can syncope [16]. Patients may also report worsening
be either symptomatic or asymptomatic, thus of symptoms during exercise, within 2 h of eat-
­creating a barrier to both recognition and treatment ing, or that symptoms are exacerbated by large,
[24]. It is the recommendation of the American carbohydrate rich meals and alcohol [16].
Autonomic Society that patients with peripheral Symptoms may show diurnal variability (worse
neuropathies known to be associated with auto- in the morning) and are influenced by hydration
nomic dysfunction, such as diabetes, be screened and ambient temperature [14, 15].
for nOH [15]. At minimum, patients should be
asked if they have symptoms, they commonly
experience within 3–5 min of standing that resolve 3.2 Initial Steps
when sitting or lying down. There are additionally
different variants of OH, and while some occur If any of the screening questions for OH are posi-
within the first 15  s of standing (initial OH), tive, orthostatic vitals should be obtained in the
delayed OH occurs after 3 min of standing [15]. clinic. If possible, have patients track their ortho-
Please see Table 1 for a full list of recommended static vital signs at home upon awakening, when
screening questions in patients with suspect nOH. symptomatic, and prior to bedtime for around a
week to aid in accurate clinical assessment of the
condition [15]. In addition to recording changes
3.1 Symptoms in blood pressure, orthostatic changes to heart
rate should be evaluated. A change in heart rate of
Orthostatic hypotension is thought to be one of <15 bpm within 3 min of moving from lying to
the most incapacitating manifestations of auto- standing in the setting of OH suggests that the
nomic failure [6]. Symptoms are limited to stand- OH is due to neurogenic causes because the com-
ing and resolve when sitting or lying down. pensatory sympathetic heart rate increase is
Inadequate perfusion of the brain may cause diz- impaired [25]. In nOH, a ΔHR/ΔSBP ratio
ziness, lightheadedness, cognitive slowing, <0.5  beats/min/mmHg is suggestive of neuro-
blurry or dimmed vision, “coat hanger” pain in genic orthostatic hypotension [25]. Conversely,
heart rate change >15 bpm or ΔHR/ΔSBP ratio
>0.5  beats/min/mmHg is suggestive of non-­
Table 1  Screening questions for patients with suspect
orthostatic hypotension neurogenic cause of orthostatic hypotension [25].
1. Have you fainted/blacked out recently?
The relevance of the heart rate ratio was evalu-
2. Do you feel dizzy or lightheaded upon standing? ated in a prospective study of 423 patients and
3. Do you have vision disturbances when standing? neurogenic OH was reliably distinguished from
4. Do you have difficulty breathing when standing? other causes of OH when the ΔHR/ΔSBP ratio at
5. Do you have leg buckling or leg weakness when 3  min of tilt was <0.5  beats/min/mmHg [25].
standing? However, this criterion can be confounded by
6. Do you ever experience neck pain or aching when medications that interfere with heart rate, intrin-
standing?
sic cardiac rhythm disturbances that impair car-
7. Do the above symptoms improve or disappear when
you sit or lay down? diac compensatory increase in heart rate, and by
8. Are the above symptoms worse in the morning or baseline variability in age and disease status [15].
after meals?
9. Have you experienced a fall recently?
10. Are there any other symptoms you commonly 3.3 Evaluation for Additional
experience when you stand up or within 3–5 min of
standing and get better when you sit or lay down? Causes of OH
Any of the questions that are answered “yes” should
prompt measurement of orthostatic vital signs In diabetic patients with changes in blood pres-
Adapted from Gibbons et al. 2017 [15] sure and heart rate suggestive of OH, history,
456 L. F. Fanty and C. H. Gibbons

physical exam, and basic diagnostics should be 3.4 Additional Autonomic Testing
obtained to evaluate for other non-neurogenic
causes of OH. An electrocardiogram can be used If clinical suspicion is high for OH and vital sign
to evaluate for cardiac dysfunction, complete testing is unrevealing, clinicians should consider
blood count to look for infection and anemia, conducting further testing to evaluate for delayed
basic metabolic panel for electrolyte distur- OH [13, 26]. These tests might include 24-h
bances, acid base disorders, volume depletion ambulatory blood pressure monitoring, supine
and renal impairment, thyroid stimulating hor- and standing norepinephrine levels, and more
mone, vitamin B12, albumin to identify chronic specialized autonomic testing such as tilt table
illness or poor nutrition, and liver enzyme testing testing, autonomic reflex testing (heart rate vari-
to evaluate for liver dysfunction [15]. Serum and ability to paced breathing, heart rate, and blood
urine protein electrophoresis can be considered if pressure response to a Valsalva maneuver), and
not already completed as part of a neuropathy sudomotor function testing [15].
workup. Neurologic antibodies can be sent rarely
if clinical suspicion is high [15]. It is important to 3.4.1 Valsalva Maneuver
review the patient’s medication list as polyphar- In the Valsalva maneuver, the patient exhales
macy is common in patients with diabetes and against resistance in order to increase intratho-
increases the risk of OH [15]. See Table 2 for a racic pressure resulting in reduced cardiac pre-
list of medication types that may cause or exacer- load. The Valsalva maneuver is done in a
bate OH. standardized manner. The patient rests in a

Table 2  Medications that may cause or worsen orthostatic hypotension


Medication class Common examples
Alcohol
Anticholinergics Atropine, glycopyrrolate, hyoscyamine
Antidepressants/tricyclics Amitriptyline, nortriptyline, imipramine, desipramine, trazodone,
selective serotonin reuptake inhibitors
Benzodiazepines Alprazolam, chlordiazepoxide, clonazepam diazepam, lorazepam
Central sympatholytic agents Centrally acting α2 agonists: clonidine
False neurotransmitters: alpha-methyldopa
Dopaminergic agents Levodopa, bromocriptine, cabergoline, apomorphine, pramipexole,
ropinirole, rotigotine
Drugs that induce autonomic neuropathy Amiodarone, vincristine, cisplatin
Insulin
Muscle relaxants Tizanidine
Negative inotropic/chronotropic agents Beta-adrenergic blockers: propranolol, metoprolol, atenolol, carvedilol,
labetalol
Non-dihydropyridine calcium channel blockers
Neuroleptics Chlorpromazine, quetiapine
Opioids (rare side effect) Oxycodone, hydrocodone, morphine, hydromorphone, heroin
Preload reducers Diuretics: acetazolamide, furosemide, hydrochlorothiazide,
spironolactone, torsemide
Nitrates: isosorbide dinitrate, nitroglycerine, nitroprusside
Phosphodiesterase 5 inhibitors: sildenafil, tadalafil, vardenafil
Renin-angiotensin system (RAS) Angiotensin converting enzyme inhibitors: captopril, enalapril, lisinopril
antagonists Angiotensin receptor blockers: losartan, telmisartan, candesartan, valsartan
Vasodilators α1 adrenoceptor antagonists: alfuzosin, doxazosin, prazosin, terazosin,
tamsulosin, carvedilol, labetalol
Dihydropyridine calcium channel blockers: amlodipine, nifedipine,
nicardipine
Other vasodilators: hydralazine, minoxidil
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 457

Fig. 1  The phases of


Phase of Valsalva
the Valsalva maneuver.
In the top panel the heart I II early II late III IV
rate is shown in a solid
black line in beats/min. 120

Heart Rate
In the middle panel the
range of systolic and 90
diastolic blood pressures
(mmHg) during the 60
Valsalva maneuver are Heart Rate
shown with the thick
grey line. In the lower
panel, the expiratory
pressure (mmHg) is
120
shown with the solid
black line
60
Blood Pressure

30
mmHg

0
Expiratory Pressure
In

Ex

En
sp

pir

d
ira

Ex
at
tio

ion

pir
n

at
ion

recumbent position and is asked to blow into a heart rate, increased cardiac output, and
peripheral vasoconstriction [27].
mouthpiece connected to a manometer and main-
• Phase III: Marks the end of strain/expira-
tain a column of mercury at 40 mmHg for 15 s tion. With release in positive intrathoracic
[27]. Beat-to-beat blood pressure recordings are pressure, blood pressure falls as there is
obtained and the four main phases of Valsalva expansion of the pulmonary vascular bed,
and heart rate increases [27].
maneuver are analyzed [see Fig.  1]. The four
• Phase IV: Characterized by an overshoot of
phases include: blood pressure above baseline. Normal
venous return of the heart coupled with
• Phase I: At the beginning of strain/expira- sympathetic activity from phase II leads to
tion, there is a transient rise in blood pres- overshoot of blood pressure from baseline.
sure and a fall in heart rate due to propulsion Increased blood pressure stimulates barore-
of blood into peripheral circulation and flex and return to normal blood pressure.
compression of the aorta [27]. Baroreflex also results in bradycardia
• Phase II: This phase has an early fall and before return to baseline [27].
then recovery of blood pressure. Positive
intrathoracic pressure leads to impaired
venous return, reduced preload and stroke In sympathetic adrenergic failure, there is a
volume, and overall fall in blood pressure. loss of phase II recovery and phase IV overshoot
The fall in blood pressure activates aortic and a delay in blood pressure recovery time. The
and carotid baroreceptors leading to
increased sympathetic discharges, increased
sensitivity and specificity of this test to detect
458 L. F. Fanty and C. H. Gibbons

adrenergic failure and separate neurogenic from caution in patients with a sensory neuropathy who
non-neurogenic OH are both 80% [27]. are at risk for ulcer formation) [15, 16, 19]. Based
on the clinical response, pharmacologic interven-
3.4.2 Tilt Table tions may be considered. Please note that it is rec-
The tilt table allows for a controlled environment ommended to wait 2  weeks between any
to monitor the autonomic response to orthostatic interventions to fully assess efficacy [17].
stress. During a tilt table test, a patient is pas-
sively transitioned to the upright position to at
least 60°. Gravity redistributes an estimated 500– 5 Medication Review
8000 mL of blood (10% of total blood volume) and Adjustment
from cerebral and thoracic vasculature to the
splanchnic and lower extremities [27]. Upon Medications that reduce intravascular volume,
transitioning to the upright posture, the force of promote vasodilation, reduce preload, or are neg-
gravity causes the blood volume to redistribute ative inotropic/chronotropic agents can exacer-
away from the cerebral and thoracic vasculature bate symptoms of OH. These medications should
toward the splanchnic and lower extremity vascu- be reviewed with patients and prescribing physi-
lature. The tilt test is a stronger orthostatic stress cians and slowly reduced/discontinued as clini-
because it removes the leg contraction that occurs cally safe. Blood pressure and heart rate responses
during standing [28]. In healthy individuals, should be monitored in response to changes to
moving to the upright position unloads the carotid help identify offending agents. Please refer to
and cardiopulmonary baroreceptors leading to a Table 2 for comprehensive list of medication cat-
reflex increase in sympathetic adrenergic output egories that should be removed if possible [15].
[27, 28]. The sympathetic adrenergic response
leads to increased peripheral vasoconstriction,
increased heart rate, and increased cardiac con- 6 Nonpharmacological
tractility. In nOH, there is an insufficient auto- Interventions
nomic response to orthostatic stress leading to a
drop in blood pressure without a compensatory 6.1 Lifestyle
tachycardia. Tilt table testing often detects OH
within 3–5 min, but up to 60 min may be required 6.1.1 Volume Repletion: Increase
to detect delayed OH [29]. Hydration and Salt Intake
Many patients are chronically volume depleted
and benefit from increasing daily water intake. A
4 Treatment minimum of 2 L daily (64 oz) is recommended to
ensure adequate hydration and if cardiac status
In patients with nOH secondary to diabetes, man- permits, up to 3  L daily is recommended [15].
agement of glycemic index should be optimized Patients with nOH can also bolus water intake
[2, 6, 30]. Further treatment of OH should be with rapid results. A 16  oz (500  mL) bolus of
geared toward optimal management of symptoms water can increase SBP by 30  mmHg in about
and not toward a specific target blood pressure. 5–10 min [15, 16, 31]. Effects however are short-­
Initial treatments should first focus on nonphar- lasting, wearing off after 45–60  min [16].
macologic measures such as removal of offending However, this can be a useful tool to counteract
medications, increase in dietary salt and fluid postprandial hypotension or OH in the setting of
intake, education on the use of physical counter-­ specific brief activities.
maneuvers, regular exercise, sleeping with head The 24 h urine sodium for patients not taking
of bed elevated (to reduce supine hypertension diuretics or fludrocortisone can be checked to
and reduce nocturia), and use of specific compres- ensure adequate daily sodium intake is occurring.
sion garments (but these must be considered with If urine sodium is <100 mEq/24 h, then consider
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 459

salt repletion. It is recommended 1–2 teaspoons 6.1.4 Purposeful Movement


of salt be added to daily diet; however, this can be Valsalva maneuvers or straining with closed glot-
increased up to 10 g for those that required vol- tis can lead to decreased preload and worsening
ume repletion. For those at risk for cardiovascu- of orthostatic symptoms and should be performed
lar complications, salt intake and clinical cautiously (e.g. fainting on the toilette is a com-
response should be monitored closely and down-­ mon situation in the setting of constipation) [15,
titrated as needed. An alternative is salt tabs 16]. Patients should change positions gradually
0.5–1 g, with the caveat they can cause abdomi- and sit before standing. Activities like leg cross-
nal discomfort [15, 16]. ing, standing on tiptoes, squatting, and buttock
clenching can all help to promote venous blood
6.1.2 Avoid Warm, Humid return [16].
Environments and Increase
in Core Body Temperature 6.1.5 Eat Small, Low Carbohydrate
Elevation in body temperature can lead to vasodi- Meals
lation and loss of intravascular volume through Orthostatic symptoms may worsen after meals
sweating, both contributing to more severe drop due to pooling of blood in the splanchnic bed.
in orthostatic blood pressure. Thus, saunas, hot To minimize this effect, recommend eating
tubs, and hot showers should be avoided. Shower small meals with low glycemic index.
chairs can be utilized for an extra safety measure Avoiding sugary beverages will also be bene-
[15, 16]. In addition, sudden exposure to hot ficial [15, 16].
humid environments (such as winter vacations to
a warm environment) can be fraught with peril 6.1.6 Avoid Alcohol and Caffeine
without adequate preparation and hydration. Alcohol can act as a vasodilator, thereby worsen-
Patients should be counseled on the need to avoid ing symptoms of nOH.  Although caffeine is a
outdoor activities during the hottest part of the stimulant, it also has diuretic properties and can
day and should take it slowly until they know lead to intravascular depletion and worsening
how their blood pressure will respond. symptoms of nOH if not consumed with appro-
priate water hydration [15, 16]. In general prac-
6.1.3 Remain Active tice, avoiding both alcohol and caffeine helps to
High intensity activity which can raise body tem- prevent complications, but can be considered in
perature should be avoided. However, regular special situations with appropriate forethought
exercise is encouraged with adequate prehydra- (this often helps to preserve the physician–patient
tion to prevent deconditioning [15]. Exercises relationship).
such as swimming, rowing, and recumbent bike
may be better tolerated than standing exercises 6.1.7 Sleep with Head of Bed
because they remove the orthostatic stress of Elevated
standing. Exercising in pools is beneficial as Vascular depletion occurs overnight secondary
hydrostatic pressure from water counteracts grav- to pressure natriuresis in the setting of supine
ity induced pooling of blood [16]. However, hypertension [16]. Sleeping with the head of
patients must take extra care when emerging bed elevated approximately 6  in. can help to
from the pool because the sudden loss of hydro- reduce nocturnal diuresis and symptomatic
static pressure can result in dramatic OH [16]. orthostatic hypotension in the morning.
Prolonged bedrest (such as during a hospitaliza- Elevating head of bed also helps to maintain
tion) can result in significant deconditioning in a renin–angiotensin–aldosterone system to main-
short period of time and dramatically worsen tain higher blood pressure in the morning [15,
OH.  Patients will require aggressive physical 16]. Additionally, sleeping with the head of bed
therapy in order to gain back the orthostatic toler- elevated helps reduce neurogenic supine
ance that is lost during deconditioning. hypertension.
460 L. F. Fanty and C. H. Gibbons

6.1.8 Compression Garments tion and correction of anemia via repletion of


Compression garments applying 30–40  mmHg iron, vitamins, or erythropoietin as indicated can
can be used to help prevent venous pooling. With help ameliorate symptomatic nOH [15, 19].
most venous pooling occurring in the splanchnic
bed, waist high compression garments or abdom-
inal binders are the most effective. A 2016 study 7.2 Fludrocortisone
found that an inflatable automated abdominal
binder which provided 40  mmHg splanchnic Although one of the most frequently prescribed
venous compression was non-inferior to mido- agents for orthostatic hypotension, fludrocorti-
drine in improving orthostatic symptoms. While sone is not approved by US Food and Drug
promising, it should be noted that sample size Administration for this indication [15]. As a syn-
was small [16, 32]. Limitations to compression thetic mineralocorticoid, fludrocortisone
garments include poor compliance due to dis- increases renal sodium and water reabsorption
comfort and difficulty putting on and ulcers in resulting in intravascular volume expansion and
patients with neuropathy impairing sensation in increased blood pressure [15, 16]. It also works
areas of compression [15, 16]. synergistically with adrenergic agonists to
enhance pressor effect [16]. Starting dose is typi-
cally 0.05–0.2 mg/day with little therapeutic ben-
7 Pharmacologic Interventions efit above 0.2–0.3  mg/day [15, 16]. Effects of
fludrocortisone can take up to 3–7 days to appre-
When nonpharmacological measures fail to ciate [15, 16]. Common side effects include
improve symptomatic nOH, pharmacological supine hypertension, hypokalemia, and ankle
intervention should be considered. There are two edema [16]. Long term use worsens hypertension
complementary strategies used: expanding intra- and can result in end-organ damage, especially
vascular volume and increasing peripheral vascu- left ventricular hypertrophy and renal failure
lar resistance [16]. Expanding intravascular [16]. This medication should be used cautiously
volume has been traditionally achieved by using in patients with congestive heart failure and is
the mineralocorticoid fludrocortisone and treat- associated with a higher risk of all cause hospital-
ment of underlying anemia, if present. Peripheral ization [15, 16].
vascular resistance can be increased with mido-
drine, droxidopa, pseudoephedrine, or norepi-
nephrine reuptake inhibitors. It is also worth 7.3 Midodrine
noting that all patients being treated for nOH
with pharmacologic agents should be wary of Midodrine has been FDA approved for the treat-
supine hypertension and so should avoid horizon- ment of OH since 1996 [16, 33]. Midodrine is a
tal positions and sleep/rest with the head of bed prodrug that is converted peripherally into its
elevated 30–45°. See Table  3 for a review of metabolite desglymidodrine, a selective alpha-1
potential therapies. adrenoceptor agonist [16]. Desglymidodrine
increases vascular resistance and blood pressure
through arteriolar and venous constriction [16].
7.1 Treatment of Anemia Midodrine is short acting lasting 1–3 h and typi-
cal dosing is 2.5–15 mg up to three times daily
Decreased blood viscosity and oxygen carrying during waking hours [16, 33]. A major side effect
capacity caused by anemia can exacerbate nOH is supine hypertension, and it is recommended
[15]. Iron deficiency anemia leads to increased that midodrine not be taken within 5 h of bedtime
nitric oxide, a potent vasodilator and possibly [15, 16]. Other side effects include piloerection,
another contributor to nOH. Therefore, investiga- scalp itching, and urinary retention [16]. Extra
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 461

Table 3  Treatment of symptomatic orthostatic hypotension and related pathology


Treatment of symptomatic orthostatic hypotension
Nonpharmacologic interventions
• Avoid medications that could worsen or cause orthostatic hypotension
• Avoid increase in core body temperature (warm and humid environments such as saunas, hot tubs, and hot
showers)
• Move with purpose:
 – Dorsiflex the feet or perform hand grip exercise before standing
 – Change positions slowly
 – Perform exercises to promote venous return such as standing on both legs and crossing them, clenching
buttocks
• Remain active
 – Trial water exercises, rowing, recumbent bike
• Elevate head of bed by 10–20°
• Increase hydration
 – 2–3 L of water daily
 – 500 mL boluses can lead to instant improvement in low blood pressure
• Increase daily sodium; 1 teaspoon daily additional to heart healthy diet; can increase Na intake to 6–10 g/daily
• Eat small, carbohydrate low meals
• Avoid alcohol as it can cause vasodilation
• Avoid coffee as diuretic properties can lead to decreased intravascular volume
• Trial abdominal compression garments
Pharmacologic interventions
Medication Mechanism of action Dose
Midodrine α-1 adrenoceptor agonist 2.5–15 mg up to three times a day during day time;
not to take within 5 h of bedtime
Droxidopa Synthetic norepinephrine precursor 100–600 mg up to three times a day during day time;
not to take within 5 h of bedtime
Fludrocortisone Synthetic mineralocorticoid 0.05–0.2 mg daily
Pyridostigmine Acetylcholinesterase inhibitor 30–60 mg up to three times daily
Atomoxetine Norepinephrine reuptake inhibitor 10–18 mg twice daily
Consideration: Treatment of anemia recommended

caution should be utilized when prescribing to Droxidopa is short acting with a peak response of
patients with congestive heart failure and chronic 3.5–6  h [15, 16]. Starting dose is 100–600  mg
renal failure [15]. three times daily during waking hours [16, 34].
Common side effects include supine hyperten-
sion, headache, nausea, dizziness, and fatigue.
7.4 Droxidopa Extra caution should be utilized when prescrib-
ing to patients with congestive heart failure and
Droxidopa has been FDA approved for the treat- chronic renal failure [15, 16].
ment of OH since 2014 [15, 34]. Droxidopa is a It is important to note that patients with a
norepinephrine prodrug that is converted to nor- supine plasma norepinephrine level <220 pg/mL
epinephrine by l-amino decarboxylase in the have a greater blood pressure response to droxi-
kidneys, the central nervous system, and in dopa treatment [16]. Palma et al. found an inverse
peripheral sympathetic postganglionic nerve end- relationship between supine plasma norepineph-
ings [15, 16]. The increase in peripheral neural rine and blood pressure increase following droxi-
norepinephrine is believed the primary mecha- dopa [2, 16]. This is explained by denervation
nism of action for increasing blood pressure. sensitivity of postganglionic adrenergic ­receptors,
462 L. F. Fanty and C. H. Gibbons

which occurs in around 25% of DAN patients, incontinence [15]. One of the benefits of the use
resulting in an exaggerated response to adrener- of pyridostigmine is it does not contribute to
gic agonists such as droxidopa and in theory, supine hypertension.
midodrine [16]. Droxidopa notionally may be
less effective in patients on carbidopa, which
blocks the conversion of droxidopa to norepi- 8 Special Considerations
nephrine [16]. There may not be all that many
patients with both diabetic autonomic neuropathy 8.1 Neurogenic Supine
and Parkinson’s disease, but in those situations Hypertension
the use of carbidopa with droxidopa should be
considered with caution. Most patients with neurogenic OH will experi-
ence supine hypertension which is defined as sys-
tolic BP ≥140  mmHg and/or diastolic BP
7.5 Norepinephrine Reuptake ≥90 mmHg [37]. All patients with supine hyper-
Inhibitors tension should be advised to avoid horizontal
supine position, including during naps, and ele-
Norepinephrine reuptake inhibitors, such as ato- vate head of bed at least 6 in. at night [5, 16]. A
moxetine, increase the availability of norepi- carbohydrate rich snack or alcoholic beverage
nephrine at the neurovascular junction. In patients right before bedtime can aide in vasodilation of
with intact central autonomic function, the effect the splanchnic bed and lower blood pressure [15,
on peripheral sympathetic postganglionic fibers 16]. Fludrocortisone should be avoided if possi-
leading to vasoconstriction is counteracted by the ble, as a main side effect is supine hypertension
vasodilatory effect of central α2 receptors [16, [15]. A consensus of expert opinion released in
35]. In patients with central autonomic dysfunc- 2017 recommends prioritizing treatment of symp-
tion, norepinephrine reuptake inhibitors result in tomatic nOH over supine hypertension as symp-
peripheral vasoconstriction without vasodilation tomatic nOH can be immediately debilitating and
from central α2 receptors [16]. This makes atom- lead to increased falls and potentially death [15].
oxetine a treatment for nOH from central auto- Short acting antihypertensives (captopril, cloni-
nomic failure seen in diseases such as multiple dine, hydralazine, losartan) may be given at night
system atrophy, but atomoxetine is less likely to time if the decision is made to treat supine hyper-
be effective for a primarily peripheral process tension. For supine SBP 160–180, the decision to
such as DAN [2, 16]. treat should be individualized. For severe supine
hypertension (SBP > 180, DBP > 110) treatment
should be initiated [17]. Patients started on anti-
7.6 Other hypertensives at night should be warned to take
extra precautions when getting out of bed at night
Pyridostigmine is used off-label for the treatment and use a urinal or bedside commode to decrease
of nOH. Pyridostigmine is a cholinesterase inhib- falls [15]. See Table 4 for a review of treatments
itor and enhances cholinergic neurotransmission for supine hypertension.
at peripheral cholinergic synapses, including
sympathetic ganglia [36]. It has been shown to
increase systolic blood pressure by only 4 mmHg 8.2 Postprandial Hypotension
and is more useful in patients whose symptoms
are less severe and when used as an adjunctive The treatment of postprandial hypotension dif-
[36]. Dosing starts at 30–60  mg once to three fers from the treatment of orthostatic hypoten-
times a day [15]. Adverse side effects include sion because of shorter anticipated periods of
cholinergic symptoms such as abdominal cramp- time where blood pressure may be low.
ing, diarrhea, sweating, sialorrhea, and urinary Nonpharmacologic measures include eating
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 463

Table 4  Treatment of supine hypertension Table 5  Treatment of postprandial hypotension


Nonpharmacologic interventions Nonpharmacologic interventions
• Carbohydrate snack or if drinking alcohol, have a • Smaller meals with lower glycemic index to reduce
glass right before bedtime splanchnic pooling
• No medications for orthostatic hypotension within • Drink 12–18 oz of water 15 min before mealtime
5 h of bedtime • Avoid sugary beverages and alcohol
• Short acting antihypertensives prior to bedtime • Monitoring of symptoms and avoidance of strenuous
• Extra precaution when getting out of bed at night if exercise 60 min after a meal
pharmacologic intervention pursued Pharmacologic interventions
Pharmacologic interventions Medication Mechanism of Dose
Medication Mechanism of Dose action
action Acarbose α-glucosidase 50–100 mg
Captopril ACE inhibitor 25 mg at night inhibitor before meals
Clonidine Central α-2 0.2 mg with Midodrine α-1 2.5–15 mg
agonist evening meal adrenoceptor before meals
Hydralazine Relaxation of 10–25 mg at agonist
peripheral night Octreotide Somatostatin 50 μg, 30 min
smooth muscles analogue before each
Losartan Angiotensin II 50 mg at night meal
receptor
antagonist
Nitroglycerin Vasodilator 0.1 mg/h patch
Patch at night to be 9 Sudomotor Dysfunction
removed in the
morning
Sudomotor function refers to the regulation of
sweating through the stimulation of eccrine sweat
small, low carbohydrate meals, throughout the glands. Sudomotor dysfunction can result in an
day and the avoidance of alcohol and sugary increase in sweat production (hyperhidrosis),
drinks [15, 16]. Patients can also drink 12–18 oz decrease in sweat production (hypohydrosis), or
of water after a meal, but before they stand up. lack of sweat production (anhidrosis). Sweating
Blood pressure is usually lowest 30–60 min after abnormalities are common in many types of neu-
a meal, and patients should monitor for symp- ropathy and sudomotor dysfunction is one of the
toms and encouraged to sit or lie down if needed. earliest detectable abnormalities in distal small
If nonpharmacological interventions fail to fiber neuropathy. The sudomotor system serves
improve symptoms, clinicians can consider phar- as the primary mechanism for evaporative heat
macologic intervention [16]. Midodrine, taken loss, and an integral part of the autonomic system
right before or during a meal can help decrease maintaining thermal homeostasis [38, 39].
venous pooling [16]. Insulin is a vasodilator, so
acarbose, a medication which reduces the gastro-
intestinal absorption of glucose, helps decrease 10 The Anatomy
postprandial hypotension by reducing insulin and Pathophysiology
release [16]. Another option is octreotide, a med- of Sudomotor Function
ication that induces vasoconstriction of splanch-
nic vessels and can attenuate postprandial The body’s autonomic system maintains an inter-
hypotension [22]. However Octreotide is admin- nal temperature around 37 °C [38]. In response to
istered subcutaneously, is often cost prohibitive, a rise in body temperature, the autonomic ner-
and can induce nausea and abdominal pain. vous system helps cool body temperature through
Midodrine, taken right before or during a meal sympathetic responses such as generalized sweat-
can help decrease venous pooling [16]. See ing, vasodilation, and hyperpnea [38]. The body
Table 5 for review of treatments for postprandial has cutaneous, visceral, and spinal cord thermo-
hypotension. receptors which initially detect an increase in
464 L. F. Fanty and C. H. Gibbons

temperature [38]. Thermal receptors and thermal occurs adjacent or distal to areas of decreased
nociceptors are typically A-delta and C fibers, sweating. In diabetic autonomic neuropathy, this
both small, unmyelinated axons [40]. These leads to increased sweating adjacent to the palms
fibers enter the spinal cord along Lissauer’s tract and soles of the feet as well as more centrally
before traveling one to two vertebral levels above over the chest and forehead [41]. Focal compen-
the level of entry to synapse on the gray matter of satory hyperhidrosis is different from primary
the dorsal horn [38]. Axons of second order neu- focal hyperhidrosis. Primary focal hyperhidrosis
rons then decussate and join the spinothalamic affects the palms, soles, and axilla and often
tract to ascend to the reticular formation, thala- occurs in response to an emotional trigger. It is
mus, and preoptic areas of the hypothalamus independent of thermoregulatory needs [38].
[38]. There have been a few cases of diabetic neu-
The hypothalamus integrates the new thermal ropathy associated with gustatory sweating,
information with references to changes in fluid another focal hyperhidrosis which presents as
volume and electrolyte concentration, resulting unilateral facial sweating in response to spicy
in thermoregulation [39]. If cooling is required, foods [42–44]. The proposed mechanism is aber-
warm sensing neurons in the medial preoptic area rant rewiring of facial parasympathetic salivary
will activate efferent sympathetic neurons. These glands to denervated sympathetic facial sweat
neurons descend ipsilaterally to synapse on the glands [38]. Diabetic neuropathy can also be
preganglionic neurons in the intermediolateral associated with nocturnal generalized diaphore-
cell column of the spinal cord [38]. The pregan- sis, although this clinical presentation is less
glionic neurons exit through the ventral root and common [38, 41]. It is important to note that
white rami T1-L2 and synapse on the paraverte- hyperhidrosis typically affects quality of life
bral sympathetic chain ganglia [38]. rather than necessitating urgent medical atten-
Postganglionic sudomotor and vasomotor fibers tion. Rarely, severe generalized hyperhidrosis
then travel to the dermis and synapse on sweat can lead to hypothermia and hypovolemia, while
glands and cutaneous arterioles to regulate evap- generalized anhidrosis can result in
orative and radiant heat loss, respectively [38]. hyperthermia.
The greatest density of thermoregulatory eccrine As mentioned above, focal compensatory
glands is on the forehead, then the upper limbs, hyperhidrosis results from underlying hypohi-
the trunk, and the lower limbs. Sweat gland den- drosis or anhidrosis that is typically in a length-­
sity is high in the palms and soles of the feet, but dependent fashion [41]. In diabetic autonomic
these glands mediate emotional sweating as well neuropathy, the most frequent pattern of sweat
as thermoregulation [38, 39]. Eccrine sweat loss is in the traditional stocking and glove distri-
glands are primarily innervated by unmyelinated bution, conforming to the pattern of length
C fibers synapsing on cholinergic M3-type recep- dependency [38]. Thoracic nerve roots may also
tors. Sympathetic sudomotor neurons can also be involved resulting in asymmetric sweat loss in
co-express markers of noradrenergic neurons, but affected areas. Progressive loss of sudomotor
parasympathetic response on sudomotor function function can result in global anhidrosis with
is thought to be negligible [38, 39]. resultant thermoregulatory impairment and
hyperthermia. At this stage, a patient normally
has profound generalized autonomic neuropathy
11 Clinical Presentation [6]. On exam, skin changes often occur due to
denervation of sweat glands, which results in tro-
Patients with diabetic autonomic neuropathy phic skin changes and hair loss. Peripheral auto-
often present with concerns of increased sweat- nomic dysfunction may also lead to observable
ing on their face and chest attributable to focal physical exam findings such as edema, rough
compensatory hyperhidrosis [41]. This form of patches of skin, loss of nails, and poor wound
hyperhidrosis is a compensatory mechanism and healing which can be contributing factors in the
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 465

development of foot ulcers. One study found the axon reflex mediated sweat response. Typical
odds ratio of foot ulcers increased to 15.3 if the testing sites include forearm proximal and distal
patient lacked sympathetic skin response [45]. leg and dorsum of foot [39]. A multi-­
compartmental capsule is placed on the skin. The
outer ring is typically filled with 10% acetylcho-
12 Diagnostics line and the inner well has nitrogen gas with a
hygrometer on the outflow tract. Iontophoresis of
Quantitative analysis of sudomotor dysfunction acetylcholine is started for 5 min and humidity is
is an important component of autonomic testing recorded from baseline to 15  min poststimula-
and can help confirm, monitor progression, and tion. Results are analyzed by assessing change in
identify treatment success of autonomic dysfunc- relative humidity and sweat latency [39, 46].
tion. Assessment of sudomotor dysfunction by Although it is the most utilized test, QSART
eliciting direct or indirect sweat response remains requires specialized equipment and is expensive
one of the most sensitive and specific means to [39]. In patients with diabetes, there is a length-­
detect small fiber neuropathy. Direct sweat dependent reduction to sweat volume beginning
response can be obtained by using iontophoresis at the foot and progressing to more proximal sites
of cholinergic agonists to stimulate M3 musca- with disease progression [20].
rinic receptors [39]. An indirect axon reflex sweat
response can also be observed with direct stimu-
lation by observation in adjacent regions. When 12.2 Quantitative Direct
cholinergic agonists bind the nicotine receptors and Indirect Axon Reflex
on sudomotor nerves, an impulse travels anti- Testing (QDIRT)
dromically up the postganglionic sudomotor
axon. When it reaches a branch point, the impulse QDIRT measures the postganglionic sudomotor
then travels orthodromically until it reaches a dif- function by quantifying direct and axon reflex
ferent population of sweat glands [38, 39, 46]. mediated sweat response. Sweat glands are stim-
Tests of sudomotor function include quantita- ulated by acetylcholine iontophoresis. Sweat
tive sudomotor axon reflex testing (QSART), glands are visualized by indicator dye and a digi-
quantitative direct and indirect axon reflex testing tal image is taken every 15 s for 7 min. The sweat
(QDIRT), silicone impressions, acetylcholine droplets are then quantified by number, size,
sweat-spot, thermoregulatory sweat testing location, and latency. As it is a relatively new
(TST), and sympathetic skin response (SSR). technique, a limitation of this technique is pau-
The choice of test to perform often is dictated by city of data on normative values [47]. In patients
resources and lesion localization [39]. It is impor- with diabetes, there is typically a reduction in
tant to note that all tests of sudomotor function sweat droplet number and sweat area in the set-
are susceptible to artifact and confounding vari- ting of neuropathy [47].
ables such as ambient temperature, hydration,
medication use, age, and gender [39].
12.3 Silicone Impressions

12.1 Quantitative Sudomotor Axon Silicone impressions measure the postganglionic


Reflex Test (QSART) sudomotor function by quantifying direct and
axon reflex mediated sweat response [38]. During
The most widely utilized test of autonomic dys- this test, sweat glands are stimulated by choliner-
function is QSART, which is thought to be a sen- gic iontophoresis [48]. A thin layer of moldable
sitive and reproducible test of sudomotor function silicone material is then placed on the skin. Sweat
[39, 46]. QSART tests the integrity of the post- droplets displace the silicone material during
ganglionic sympathetic neuron by measuring the polymerization resulting in impressions that can
466 L. F. Fanty and C. H. Gibbons

be quantified [39]. Data is reported as droplet lower pH of sweat [38]. Heat and humidity are
number, size, distribution, and volume (which increased so the patient’s oral temperature
can be estimated by assuming droplets form a increases by 1.0 °C above their baseline tempera-
hemisphere) [38]. When interpreting results, it is ture. Digital photographs help generate a sweat
important to know that this testing is prone to density map [38]. Results are expressed as a
artifact as abnormal impressions can be caused TST% (measured area of anterior body anhidro-
by hair, dirt, and air bubbles and the reader may sis divided by area of anatomic figure, multiplied
not be able to differentiate between actual and by 100) [39]. The patterns of sweating are easily
artifactual impressions [39]. While it is one of the noted and TST% can provide a severity index,
easier tests to conduct, changes in moldable but also track severity of disease. It also allows
material (with more rapid polymerization) have evaluation of areas often not assessed by other
led to reduced ability for droplets to leave inden- methods such as the trunk, head, fingers, and toes
tations [39]. Silicone impressions have not been [39]. However, TST cannot differentiate between
systematically evaluated in patients with diabetic preganglionic and postganglionic lesions [50].
neuropathy. For this reason TST is often combined with tests
which evaluate for postganglionic lesions such as
QSART, QDIRT, and silicone impressions. TST
12.4 Acetylcholine Sweat-Spot is time-consuming, requires specialized equip-
ment, and is often poorly reimbursed. In patients
This test is used to evaluate postganglionic sudo- with diabetes, TST results often provide a graphi-
motor function by measuring axon reflex medi- cal depiction of a distribution of neuropathy, with
ated sweat response [39]. To perform this test, the reduced sweat production distally [38, 49].
skin is first painted with an indicator dye. An
intradermal injection of acetylcholine is then
used to stimulate sweat glands. Sweat spots can 12.6 Sympathetic Skin Response
be visualized from the indicator dye and a picture (SSR)
is taken 6 min after injection and quantified on a
grid [39]. Limitations to this method are that it SSR, also known as galvanic skin response, pro-
represents a single point in time and that some vides a surrogate measure of sympathetic cholin-
patients report the injection to be painful [39]. ergic sudomotor function through the measure of
The acetylcholine sweat-spot test also has not electrodermal activity. Recording electrodes are
been studied in detail in patients with diabetic placed on the dorsal and ventral hand and foot
neuropathy. [51, 52]. Changes in skin potential are triggered
by inspiratory gasp, forceful expiration, startle
response, or electrical stimulation and recordings
12.5 Thermoregulatory Sweat are taken via EMG [49, 50]. SSRs are reported to
Testing (TST) be absent or present; amplitude and latency are
also observed [49, 50]. This method is easy to
TST interrogates the sympathetic sudomotor perform but is not thought to be a reliable mea-
pathway from the central nervous system to the sure of sudomotor dysfunction. There is high
cutaneous sweat gland. During this test, the variability within and between subjects, and
patient is placed in a temperature and humidity SSRs decline with age and may not be seen in
controlled chamber with ceiling mounted infra- patients older than 50 years. Further, SSR is only
red heaters controlled by probes measuring the a surrogate measure of sweat gland function and
patient’s temperature on the head, trunk, and patients with congenital absence of sweat glands
proximal leg. There is also an oral cutaneous will still have an SSR response, thus creating a
probe [38, 49]. The patient is covered in an indi- clear discrepancy with other measures of true
cator dye which changes color in response to the sudomotor function [49, 50]. The sympathetic
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 467

skin response (SSR) evaluates the momentary assessment, diagnosis, and management. Diabetes
Metab Res Rev. 2011;27:639.
change in the electrical potential associated with 5. Vinik AI, Casellini C, Parson HK, Colberg SR,
induced sweating in the palms and soles. The Nevoret ML.  Cardiac autonomic neuropathy in dia-
response can be evoked by psychological or local betes: a predictor of cardiometabolic events. Front
nociceptive stimuli [53]. As the response is emo- Neurosci. 2018;12:591.
6. Freeman R.  Diabetic autonomic neuropathy. Handb
tionally activated rather than thermoregulatory, it Clin Neurol. 2014;126:63–79.
is highly variable and has limited sensitivity and 7. Soedamah-Muthu SS, Chaturvedi N, Witte DR,
specificity in the diagnosis of sudomotor nerve Stevens LK, Porta M, Fuller JH.  Relationship
impairment. SSR is a sensitive, but not specific, between risk factors and mortality in type 1 diabetic
patients in Europe: the EURODIAB Prospective
measure of sudomotor dysfunction in patients Complications Study (PCS). Diabetes Care.
with diabetes. Abnormalities to both amplitude 2008;31:1360–6.
and latency have been detected across a range of 8. Ziegler D, Zentai CP, Perz S, et al. Prediction of mor-
diabetic neuropathy [51]. However, SSR ampli- tality using measures of cardiac autonomic dysfunc-
tion in the diabetic and nondiabetic population: the
tude and latency measures do not seem to accu- MONICA/KORA Augsburg Cohort Study. Diabetes
rately estimate the severity of a neuropathy and Care. 2008;31:556–61.
cannot be used to track longitudinal changes 9. Astrup AS, Tarnow L, Rossing P, Hansen BV, Hilsted
[54]. J, Parving HH.  Cardiac autonomic neuropathy pre-
dicts cardiovascular morbidity and mortality in type 1
diabetic patients with diabetic nephropathy. Diabetes
Care. 2006;29:334–9.
13 Treatment 10. Kaur D, Tiwana H, Stino A, Sandroni P. Autonomic
neuropathies. Muscle Nerve. 2021;63:10–21.
11. Dimitropoulos G, Tahrani AA, Stevens MJ.  Cardiac
Therapy for sudomotor dysfunction related to autonomic neuropathy in patients with diabetes mel-
diabetic autonomic neuropathy primarily con- litus. World J Diabetes. 2014;5:17–39.
sists of education and lifestyle modifications. 12. Maser RE, Lenhard MJ.  Cardiovascular autonomic
Although patients’ primary concern may be neuropathy due to diabetes mellitus: clinical mani-
festations, consequences, and treatment. J Clin
proximal compensatory sweating, clinicians Endocrinol Metab. 2005;90:5896–903.
should avoid pharmaceutical treatment as this 13. Gibbons CH, Freeman R.  Clinical implications of
can increase the risk of hyperthermia. Patients delayed orthostatic hypotension: a 10-year follow-up
should be counseled on lifestyle modifications study. Neurology. 2015;85:1362.
14. Freeman R, Wieling W, Axelrod FB, et al. Consensus
such as wearing cooling garments, carrying cool- statement on the definition of orthostatic hypoten-
ant packs and spray water bottles, and avoidance sion, neurally mediated syncope and the postural
of warm environments when possible. Patients tachycardia syndrome. Auton Neurosci Basic Clin.
should also partake in careful foot care to prevent 2011;161:46–8.
15. Gibbons CH, Schmidt P, Biaggioni I, et al. The rec-
ulceration and infection. ommendations of a consensus panel for the screening,
diagnosis, and treatment of neurogenic orthostatic
hypotension and associated supine hypertension. J
References Neurol. 2017;264:1567–82.
16. Palma JA, Kaufmann H.  Management of ortho-
static hypotension. Continuum (Minneap Minn).
1. Eid S, Sas KM, Abcouwer SF, et  al. New insights
2020;26:154–77.
into the mechanisms of diabetic complications:
17. Freeman R. Clinical practice. Neurogenic orthostatic
role of lipids and lipid metabolism. Diabetologia.
hypotension. N Engl J Med. 2008;358:615–24.
2019;62:1539–49.
18. Low PA, Singer W.  Management of neurogenic
2. Sasaki H, Kawamura N, Dyck PJ, Dyck PJB, Kihara
orthostatic hypotension: an update. Lancet Neurol.
M, Low PA.  Spectrum of diabetic neuropathies.
2008;7:451–8.
Diabetol Int. 2020;11:87–96.
19. Freeman R, Abuzinadah AR, Gibbons C, Jones
3. Vinik AI, Erbas T, Casellini CM.  Diabetic cardiac
P, Miglis MG, Sinn DI.  Orthostatic hypotension:
autonomic neuropathy, inflammation and cardiovas-
JACC state-of-the-art review. J Am Coll Cardiol.
cular disease. J Diabetes Investig. 2013;4:4–18.
2018;72:1294–309.
4. Spallone V, Ziegler D, Freeman R, et al. Cardiovascular
20. Low PA, Benrud-Larson LM, Sletten DM, et  al.
autonomic neuropathy in diabetes: clinical impact,
Autonomic symptoms and diabetic neuropa-
468 L. F. Fanty and C. H. Gibbons

thy: a population-­ based study. Diabetes Care. of orthostatic hypotension in autonomic failure.
2004;27:2942–7. Hypertension. 2014;64:1235–40.
21. Dyck PJ, Kratz KM, Karnes JL, et al. The prevalence 36. Singer W, Sandroni P, Opfer-Gehrking TL, et  al.
by staged severity of various types of diabetic neurop- Pyridostigmine treatment trial in neurogenic ortho-
athy, retinopathy, and nephropathy in a population-­ static hypotension. Arch Neurol. 2006;63:513–8.
based cohort: the Rochester Diabetic Neuropathy 37. Fanciulli A, Jordan J, Biaggioni I, et  al. Consensus
Study. Neurology. 1993;43:817–24. Published erra- statement on the definition of neurogenic supine
tum appears in Neurology 1993;43(11):2345. hypertension in cardiovascular autonomic failure
22. Jansen RW, Lipsitz LA.  Postprandial hypoten- by the American Autonomic Society (AAS) and the
sion: epidemiology, pathophysiology, and clinical European Federation of Autonomic Societies (EFAS):
management. Ann Intern Med. 1995;122:286–95. Endorsed by the European Academy of Neurology
Review. (EAN) and the European Society of Hypertension
23. Gannon J, Claffey P, Laird E, Newman L, Kenny RA, (ESH). Clin Auton Res. 2018;28:355–62.
Briggs R.  The cross-sectional association between 38. Coon EA, Cheshire WP Jr. Sweating disorders.
diabetes and orthostatic hypotension in community-­ Continuum (Minneap Minn). 2020;26:116–37.
dwelling older people. Diabet Med. 2019;37:1299. 39. Illigens BM, Gibbons CH. Sweat testing to evaluate
24. Freeman R, Illigens BMW, Lapusca R, et al. Symptom autonomic function. Clin Auton Res. 2009;19:79–87.
recognition is impaired in patients with orthostatic 40. Mandadi S, Roufogalis BD.  ThermoTRP channels
hypotension. Hypertension. 2020;75:1325–32. in nociceptors: taking a lead from capsaicin receptor
25. Norcliffe-Kaufmann L, Kaufmann H, Palma JA, et al. TRPV1. Curr Neuropharmacol. 2008;6:21–38.
Orthostatic heart rate changes in patients with auto- 41. Stewart JD.  Sweating abnormalities and other auto-
nomic failure caused by neurodegenerative synucle- nomic disorders in diabetes mellitus. Mayo Clin Proc.
inopathies. Ann Neurol. 2018;83:522–31. 1989;64:712–5.
26. Gibbons CH, Freeman R. Delayed orthostatic hypo- 42. Blair DI, Sagel J, Taylor I. Diabetic gustatory sweat-
tension: a frequent cause of orthostatic intolerance. ing. South Med J. 2002;95:360–2.
Neurology. 2006;67:28–32. 43. Urman JD, Bobrove AM.  Diabetic gustatory sweat-
27. Cheshire WP, Freeman R, Gibbons CH, et  al. ing successfully treated with topical glycopyrrolate:
Electrodiagnostic assessment of the autonomic ner- report of a case and review of the literature. Arch
vous system: a consensus statement endorsed by the Intern Med. 1999;159:877–8.
American Autonomic Society, American Academy 44. Shaw JE, Parker R, Hollis S, Gokal R, Boulton
of Neurology, and the International Federation AJ.  Gustatory sweating in diabetes mellitus. Diabet
of Clinical Neurophysiology. Clin Neurophysiol. Med. 1996;13:1033–7.
2020;132:666. 45. Tentolouris N, Marinou K, Kokotis P, Karanti A,
28. Illigens BMW, Gibbons CH.  Autonomic test- Diakoumopoulou E, Katsilambros N. Sudomotor dys-
ing, methods and techniques. Handb Clin Neurol. function is associated with foot ulceration in diabetes.
2019;160:419–33. Diabet Med. 2009;26:302–5.
29. Gibbons CH, Freeman R. Delayed orthostatic hypoten- 46. Low PA, Caskey PE, Tuck RR, Fealey RD, Dyck
sion. Auton Neurosci Basic Clin. 2020;229:102724. PJ.  Quantitative sudomotor axon reflex test in
30. Andersen ST, Witte DR, Fleischer J, et al. Risk factors normal and neuropathic subjects. Ann Neurol.
for the presence and progression of cardiovascular 1983;14:573–80.
autonomic neuropathy in type 2 diabetes: addition-­ 47. Gibbons CH, Illigens BM, Centi J, Freeman
Denmark. Diabetes Care. 2018;41:2586–94. R.  QDIRT: quantitative direct and indirect test of
31. Mathias CJ, Young TM.  Water drinking in the man- sudomotor function. Neurology. 2008;70:2299–304.
agement of orthostatic intolerance due to orthostatic 48. Vilches JJ, Navarro X. New silicones for the evalua-
hypotension, vasovagal syncope and the postural tion of sudomotor function with the impression mold
tachycardia syndrome. Eur J Neurol. 2004;11:613–9. technique. Clin Auton Res. 2002;12:20–3.
32. Okamoto LE, Diedrich A, Baudenbacher FJ, et  al. 49. Fealey RD, Low PA, Thomas JE.  Thermoregulatory
Efficacy of servo-controlled splanchnic venous sweating abnormalities in diabetes mellitus. Mayo
compression in the treatment of orthostatic hypo- Clin Proc. 1989;64:617–28.
tension: a randomized comparison with midodrine. 50. Hilz MJ, Dutsch M. Quantitative studies of autonomic
Hypertension. 2016;68:418–26. function. Muscle Nerve. 2006;33:6–20.
33. McClellan KJ, Wiseman LR, Wilde MI.  Midodrine. 51. Braune HJ, Horter C.  Sympathetic skin response in
A review of its therapeutic use in the management of diabetic neuropathy: a prospective clinical and neu-
orthostatic hypotension. Drugs Aging. 1998;12:76– rophysiological trial on 100 patients. J Neurol Sci.
86. Review. 1996;138:120–4.
34. Kaufmann H, Norcliffe-Kaufmann L, Palma 52. Levy DM, Reid G, Rowley DA, Abraham
JA. Droxidopa in neurogenic orthostatic hypotension. RR.  Quantitative measures of sympathetic skin
Expert Rev Cardiovasc Ther. 2015;13:875–91. response in diabetes: relation to sudomotor and neu-
35. Ramirez CE, Okamoto LE, Arnold AC, et al. Efficacy rological function. J Neurol Neurosurg Psychiatry.
of atomoxetine versus midodrine for the treatment 1992;55:902–8.
Orthostatic Hypotension and Sudomotor Dysfunction in Diabetes 469

53. Shahani BT, Halperin JJ, Boulu P, Cohen 54. Vetrugno R, Liguori R, Cortelli P, Montagna
J.  Sympathetic skin response--a method of assess- P.  Sympathetic skin response: basic mecha-
ing unmyelinated axon dysfunction in periph- nisms and clinical applications. Clin Auton Res.
eral neuropathies. J Neurol Neurosurg Psychiatry. 2003;13:256–70.
1984;47:536–42.
Gastrointestinal Neuropathy

Karen L. Jones, Chinmay S. Marathe, Tongzhi Wu,


Christopher K. Rayner, and Michael Horowitz

1 Introduction Disordered gastric emptying (delayed and accel-


erated) has important implications for the optimi-
Gastrointestinal neuropathy is now appreciated sation of glycaemic control in diabetes.
as of major importance in patients with type 1 or Gastrointestinal autonomic impairment may also
type 2 diabetes and to have diverse clinical mani- impact the regulation of postprandial blood pres-
festations. Disordered gastric motor function is sure. The outcomes of current approaches to the
the best characterised manifestation, but auto- management of gastrointestinal autonomic neu-
nomic neuropathy can affect all regions of the ropathy are, unfortunately for the main part, sub-­
gastrointestinal tract and may be associated with optimal, partly because the underlying
heterogeneous symptoms, including heartburn, pathophysiology is still poorly defined.
nausea and vomiting, postprandial fullness, diar- This chapter addresses the epidemiology,
rhoea, constipation, and faecal incontinence. pathophysiology, and management of gastroin-
testinal autonomic neuropathy, including the
impact of gastrointestinal function on glycaemic
K. L. Jones (*) · C. S. Marathe · T. Wu ·
M. Horowitz control in type 1 and type 2 diabetes.
Adelaide Medical School, The University of
Adelaide, Adelaide, Australia
Centre of Research Excellence in Translating 2 Assessment
Nutritional Science to Good Health, The University of Gastrointestinal
of Adelaide, Adelaide, Australia Symptoms and Autonomic
Endocrine and Metabolic Unit, Royal Adelaide Function in Diabetes
Hospital, Adelaide, Australia
e-mail: karen.jones@adelaide.edu.au; chinmay.
marathe@adelaide.edu.au; tongzhi.wu@adelaide.edu.au; While gastrointestinal symptoms in diabetes are
michael.horowitz@adelaide.edu.au frequently classified in an organ-specific manner,
C. K. Rayner i.e. ‘oesophageal’ (heartburn, dysphagia). ‘gas-
Adelaide Medical School, The University of tric’ (nausea, vomiting, postprandial fullness),
Adelaide, Adelaide, Australia and ‘intestinal’ (constipation, diarrhoea, faecal
Centre of Research Excellence in Translating incontinence), there is, in many cases, substantial
Nutritional Science to Good Health, The University overlap so that the attribution of a particular
of Adelaide, Adelaide, Australia symptom to a specific region of the gastrointesti-
Department of Gastroenterology and Hepatology, nal tract often lacks validity. Moreover, gastroin-
Royal Adelaide Hospital, Adelaide, Australia testinal symptoms are also prevalent in the
e-mail: chris.rayner@adelaide.edu.au

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 471
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_27
472 K. L. Jones et al.

general population (particularly women and indi- dated, cardiovascular reflex tests are used widely
viduals who are obese), may not be volunteered as a surrogate but, by definition, are primarily an
(particularly when regarded as embarrassing), index of cardiovascular, rather than gastrointesti-
there is substantial symptom ‘turnover’ (i.e. with nal, function [6].
time, symptoms are both ‘gained’ and ‘lost’) and
symptoms are associated with both psychological
dysfunction and depression [1]. 3 Oesophagus
An important issue is that symptoms should, if
possible, not be evaluated using the so-called While the oesophagus has received much less
patient self-report, as this is known to be attention than the stomach, it is clear that both
­unreliable [1]; rather, a validated symptom ques- disordered oesophageal motility and ‘oesopha-
tionnaire should be used, as is now mandated by geal symptoms’, e.g. dysphagia and gastroesoph-
regulatory bodies for clinical trials of functional ageal reflux, occur frequently, particularly in
gastrointestinal disorders, e.g. irritable bowel patients with evidence of autonomic neuropathy.
syndrome and functional dyspepsia. A major lim- Gastroesophageal reflux is also, not infrequently,
itation in the majority of studies, which compro- asymptomatic, even when associated with ero-
mises their interpretation, has been the use of sive disease [7].
‘self-report’ to evaluate symptoms, despite the Oesophageal transit is delayed in ~50%
availability of these validated instruments, some patients with longstanding diabetes [8, 9] and
specific to diabetes [1–3], including the Diabetes associated weakly with symptoms of reflux and
Bowel Symptom Questionnaire [3] and a diary, dysphagia. The prevalence of oesophageal symp-
the Gastroparesis Cardinal Symptom Index toms in patients with diabetes is highly dependent
(GSCI-DD) [4]. This issue applies, in particular, on the population studied. A higher prevalence of
to glucose-lowering drugs that are clearly associ- symptoms of reflux has been observed in tertiary
ated with adverse gastrointestinal effects, most referral centres (~40%), than in the community
recently, glucagon-like peptide-1 (GLP-1) recep- (~15%). Symptoms of dysphagia occur in ~25%
tor agonists (RAs) [5]. The use of self-report also of patients with diabetes seen in tertiary referral
increases the potential for ‘precebo’ (expectation centres, while community-­based studies report a
of benefit) and ‘nocebo’ (expectation of harm) prevalence of 5% [10]. Whether diabetes is asso-
effects. ciated with an increased risk of Barrett’s oesopha-
Despite this limitation, it is clear that gastroin- gus remains contentious [11].
testinal symptoms occur more frequently in both
type 1 and type 2 diabetes, affect quality of life
adversely, ‘turnover’, and are associated with 3.1 Diagnosis
depression [1, 2], the use of specific glucose-­
lowering medications [1], and poor, chronic gly- Endoscopy is required to identify mucosal dis-
caemic control [1]. ease. As in non-diabetic patients, oesophageal
It should also be appreciated that the frequent motility can be assessed by conventional or high-­
attribution of gastrointestinal symptoms to auto- resolution manometry [12], to evaluate lumen-­
nomic dysfunction in diabetes is essentially one occlusive pressure waves, and contrast video
of exclusion, because of the lack of diagnostic swallow radiology demonstrates both structural
tests that can be used to specifically evaluate gas- and functional disorders. Scintigraphy is less
trointestinal autonomic function in humans. invasive and can be used to measure oesophageal
Furthermore, these tests are all indirect, e.g. the transit of swallowed radiolabelled food [8], but
plasma pancreatic polypeptide response to sham the technique lacks standardisation and is not
feeding [1, 2] and the postprandial increase in routinely employed clinically. Oesophageal pH
superior mesenteric blood flow as measured by and impedance measurements may be useful in
Doppler ultrasound [1, 2]. Non-invasive, vali- the assessment of gastroesophageal reflux.
Gastrointestinal Neuropathy 473

3.2 Pathophysiology in refuting the dogma that symptoms were the


outcome of delay in gastric emptying. Earlier
The pathogenesis of oesophageal dysfunction is Rundles (in 1945) [21] suggested that gastropa-
likely to be similar to that of gastroparesis (to be resis represented a manifestation of autonomic
discussed), with evidence of vagal dysfunction neuropathy. Diabetes is now recognised as the
[13], although the relationship of oesophageal major cause of chronic gastroparesis; abnor-
transit with the rate of gastric emptying is rela- mally delayed, and more rapid, gastric emptying
tively weak [6]. Oesophageal motility has also both occur frequently in type 1 and type 2 diabe-
been shown to be affected by acute elevations in tes [6, 22]. While it has been suggested that a
blood glucose [13, 14]. Reported motor abnor- diagnosis of gastroparesis should be dependent
malities include reductions in the number and on the concomitant presence of upper gastroin-
amplitude of peristaltic waves [15] and increased testinal symptoms, such as nausea and vomiting,
stiffness of the oesophageal body [16]. There is as well as delayed gastric emptying. We consider
also evidence that changes in central processing this inappropriate, particularly given that (as will
may contribute to symptoms [17]. be discussed) not infrequently, patients with
markedly delayed gastric emptying have few, or
no, symptoms, as Kassander initially observed
3.3 Treatment [20] and delayed emptying, even when asymp-
tomatic, has substantial clinical implications,
The management of disordered oesophageal particularly in relation to the management of
function in diabetes is for the main part empiri- glycaemia.
cal. Gastroesophageal reflux is usually treated In health, gastric emptying of solid and liquid
with proton pump inhibitors [18] and, in some nutrients exhibits a wide inter-individual, but
cases, elevating the head-end of the bed by much lower intra-individual, variation, approxi-
30–45° may prove helpful [19]. Patients with mating an overall linear rate (in the case of solids,
delayed oesophageal transit, particularly the following an initial lag phase), ranging between 1
elderly, are at risk for ‘pill-induced oesophagitis’ and 4 kcal/min [23]. Initial studies performed in
and should be advised to always drink water the 1980s, which capitalised on the development
immediately after taking oral medications. The of scintigraphic techniques to measure gastric
use of pharmacological options other than proton emptying around that time, demonstrated that
pump inhibitors, including prokinetic drugs, gastric emptying is delayed in 30–50% of indi-
lacks an evidence base. Patients with diabetes, viduals with longstanding (and then, usually
particularly when poorly controlled, have an poorly controlled) type 1 or type 2 diabetes [6].
increased propensity for oesophageal candidia- Accordingly, gastroparesis became a common,
sis, which may present as odynophagia, or be rather than a, hitherto assumed, relatively rare,
asymptomatic. disorder in diabetes [24]. A recent analysis of
DCCT-EDIC data, where gastric emptying was
measured using a stable isotope breath test,
4 Stomach reported a comparable prevalence of gastropare-
sis of 47% [25]. These studies also established
Disordered gastric emptying is the most widely that, in cases of gastroparesis, the magnitude of
recognised manifestation of gastrointestinal the delay in gastric emptying, while significant,
autonomic neuropathy in diabetes. Kassander, in is often modest, the relationship of rates of gas-
1958 [20], reported markedly increased, and also tric emptying of solids and liquids is relatively
asymptomatic, intragastric retention of barium weak, gastric emptying is occasionally acceler-
in six insulin-treated patients (5 with type 1 dia- ated, gastric emptying is reproducible in the
betes) and coined the descriptive term ‘gastropa- longer-­term (up to 25  years) and even marked
resis diabeticorum’ [20]. His report was prescient delay in emptying may not be associated with a
474 K. L. Jones et al.

poor prognosis [26]. Healthcare costs and hospi- ents to the small intestine and the action of
talisation related to diabetic gastroparesis have exogenous insulin. Kassander observed in 1958
continued to increase in the USA [27], but that that ‘the retention of stomach contents in a dia-
this may, at least in part, reflect increased aware- betic obviously may cause confusion as far as
ness. In contrast to longstanding, complicated food intake and utilisation are concerned’ [20].
diabetes, it has been appreciated more recently Gastric emptying, even variations within the nor-
that in both well-controlled type 2 diabetes [22] mal range, may affect both the timing and dosage
and adolescents with type 1 diabetes [28], gastric of insulin. As early as 1994 [37], it was reported
emptying is usually normal or mildly accelerated. that the insulin requirement to maintain eugly-
It has been suggested, probably correctly, that the caemia in type 1 patients with gastroparesis com-
latter may predispose to the development of type pared to those with normal emptying was less
2 diabetes [22, 29], a concept supported by the between 0 and 120 min and greater between 180
demonstration that in some ethnic populations and 240 min after a meal [37]. In insulin-treated
predisposed to diabetes, gastric emptying is faster patients, unexplained episodes of hypoglycae-
than in Caucasians [30, 31]. mia, particularly postprandially, may be the sole
The relationship of upper gastrointestinal feature of gastroparesis [38], the so-called gastric
symptoms, such as nausea, vomiting, and post- hypoglycaemia [39].
prandial fullness, with the rate of gastric empty-
ing and the intragastric distribution of a meal [32]
are statistically significant but, in general, weak 4.1 Diagnosis
[6]. It has recently been reported that symptoms
occurring during measurement of gastric empty- Because the prevalence of upper gastrointestinal
ing, but not the rate of gastric emptying per se, symptoms is not strongly predictive of gastropa-
are predictive of daily symptoms [33]. Of the resis, and gastroparesis may be asymptomatic,
symptoms, early satiety and fullness appear to be the diagnosis is dependent on measurement of
most strongly associated with delayed gastric gastric emptying [24]. Gastroduodenal pathol-
emptying [34]. There is evidence that vomiting is ogy, particularly gastric outlet obstruction, must
more prominent and abdominal pain less in always be excluded by upper gastrointestinal
patients with diabetic gastroparesis than is the endoscopy. A number of methods are available to
case in patients with idiopathic gastroparesis measure gastric emptying (Table 1). Irrespective
[35]. Accordingly, it is clear that delay in gastric of the choice, attempts should be made to cease
emptying is best regarded as a marker of gastro- medications that may slow gastric emptying (e.g.
duodenal motor/sensory abnormality, rather than opioids [40] and both short- [41] and long-acting
as a direct cause of symptoms, as was previously [42] GLP-1 agonists) and maintain blood glucose
believed. in the range of 4–10 mmol/L during the measure-
Despite a high prevalence of obesity, particu- ment [43]. Metformin appears to slow gastric
larly in type 2, but increasingly also in type 1, emptying, but only modestly [44]. Scintigraphy,
diabetes, malnutrition occurs frequently in
patients with gastroparesis. Gastroparesis may
Table 1  Methods to assess gastric emptying
also impact on the absorption of oral medica-
• Scintigraphy
tions, which is of particular relevance to drugs
• Stable isotope breath tests
which have a short half-life, or where a rapid • Ultrasound
onset of action is needed, as is the case with some • SPECT
oral hypoglycaemic agents [36]. In insulin- • MRI
treated patients (i.e. type 1 or insulin-treated type • Absorption kinetics of oral drugs, e.g. paracetamol
2), delayed gastric emptying has the potential to (acetaminophen)
affect glycaemic control adversely by predispos- • Radiology (barium studies/radiopaque markers)
ing to a mismatch between the delivery of nutri- • Smart Pill™
Gastrointestinal Neuropathy 475

the use of radiolabelled meals and a gamma cam- kinetics of oral paracetamol absorption have been
era, remains the ‘gold standard’ technique and used widely, particularly by the pharmaceutical
allows measurement of emptying of both solid industry, as a simple surrogate marker of gastric
and liquid meal components, potentially concur- emptying, but unfortunately, this technique has
rently [45] as well as (in research studies) intra- major limitations [51, 52] and, from our perspec-
gastric meal distribution. There have been recent tive, should not be used for clinical (and ideally
steps towards standardisation of methodology for not for research) purposes.
scintigraphic measurement, internationally [43].
The major limitations of scintigraphy are the
necessity for expensive equipment and the asso- 4.2 Pathophysiology
ciated radiation burden. Stable isotope breath
tests are now well validated and used increas- While the pathophysiology of diabetic gastropa-
ingly for measurement of gastric emptying [46, resis is complex, there have been substantial
47] with the advantages of simplicity and avoid- insights in this area in the last ~10 to 15 years,
ance of radiation exposure [46]. The methodol- particularly reflecting outcomes of the work of
ogy used to evaluate breath test data remains the Gastroparesis Clinical Research Consortium
inconsistent and, in general, gastric emptying in the USA [53]. Earlier studies demonstrating an
times are best regarded as notional, rather than association of delayed gastric emptying with an
precise, although correction using the so-called impaired pancreatic polypeptide response to
Wagner-Nelson method may generate data com- sham feeding [54] supported the concept that
parable to that of scintigraphy [48]. A further gastroparesis is a sequela of autonomic neuropa-
limitation is that the effects of gastric emptying thy. However, the relationship of gastric empty-
and small intestinal absorption of the label can- ing, measured with scintigraphy, with
not be discriminated. The wireless motility cap- cardiovascular autonomic function was shown to
sule, or Smart Pill™, can evaluate gastric be relatively weak [6] (Fig.  1). In both patients
emptying [49], but because it is a non-digestible with diabetes and rodent models, reductions in
solid, it is only emptied from the stomach after cells in the vagal motor and sensory sympathetic
emptying of digestible solids and nutrient liquids ganglia as well as structural abnormalities of
and is also insensitive in the identification of vagal nerve fibres, both intrinsic and extrinsic to
rapid emptying [50]. The plasma absorption the gut wall, have been reported [55]. In patients

Fig. 1 Relationship 120


between solid gastric
emptying (intragastric r = 0.35, p < 0.01
100
Solid retention at 100 min (%)

retention at 100 min),
measured with
scintigraphy, and 80
autonomic function
assessed by
cardiovascular reflexes 60
in unselected patients
with longstanding type 1
40
and type 2 diabetes
(n = 87). The
relationship is 20
significant but weak [6]
0

0 1 2 3 4 5 6
Score for autonomic nerve function
476 K. L. Jones et al.

with gastroparesis, a spectrum of abnormalities around smooth muscle cells and nerves has been
in gastroduodenal motor/sensory function have noted to be characteristically thickened [58]. The
been demonstrated, including impaired accom- cause of the loss of ICCs, which is now regarded
modation of the proximal stomach (i.e. the relax- as the ‘dominant’ abnormality in diabetic gastro-
ation of the stomach in response to meal paresis, remains uncertain but may be secondary
ingestion), antral hypomotility, increased con- to changes in immune function with a shift from
tractile activity localised to the pylorus, abnor- protective M2 to, activated, M1 macrophages,
malities of the gastric electrical rhythm, and reductions in insulin or IGF-1 signalling and the
‘hypersensitivity’ to both small intestinal nutri- production of stem cell factor [54], and impaired
ents and gastric distension [36, 56, 57]. The full-­ regulation of heme-oxygenase-1, resulting in
thickness gastric biopsies obtained by the increased oxidative stress [60, 61]. The expres-
Gastroparesis Clinical Research Consortium, sion of the Ano-1 gene, expressed in the ICCs
often at the time of gastric pacemaker insertion and which encodes a calcium-activated chloride
(to be discussed), have shown abnormalities in channel, may also be abnormal [62]. Abnormal
>80% of cases of severe gastroparesis resistant to duodenal mucosal mitochondrial gene expres-
conventional therapy, which are, not surprisingly, sion has recently been reported to be associated
heterogeneous. The dominant abnormalities, in with a delay in gastric emptying [63], suggesting
addition to changes in vagal innervation, are loss that mitochondrial function may be important.
or damage to the Interstitial Cells of Cajal (ICC), While it should be appreciated that the studies of
which are integral to the function of the gastric the Gastroparesis Clinical Research Consortium
‘pacemaker’ [58], an immune infiltrate in the relate primarily to patients with severe gastropa-
myenteric plexus, a reduction in intrinsic nerves resis, the heterogeneous nature of the abnormali-
and loss of inhibitory neurons expressing nitric ties demonstrated has substantial implications for
oxide synthase (nNOS) (Fig. 2). The basal lamina effective management.

a b

Fig. 2  Haematoxylin and eosin staining of the inter-­ myenteric plexus (original magnification 340) in a patient
myenteric plexus from patients with gastroparesis. (a) with severe diabetic gastroparesis [59]. (Figure courtesy
Normal ganglia and nerve fibres (original magnification of Prof H Parkman)
340). (b) Moderate lymphocytic infiltrate in the inter-­
Gastrointestinal Neuropathy 477

4.3 Effects of the Glycaemic but significant, effect to slow gastric emptying of


Environment on Gastric both solids and liquids in longstanding type 1
Emptying diabetes, measured using scintigraphy in 1990
[65] and, subsequently, elevations in blood glu-
Acute changes in the blood glucose concentra- cose within the normal postprandial range were
tion represent a reversible determinant of the rate shown to delay gastric emptying modestly in
of gastric emptying in diabetes [64–66], so that uncomplicated type 1 diabetes [64]. Acute eleva-
gastric emptying is slowed by hyperglycaemia tions in blood glucose were also shown to attenu-
and accelerated by insulin-induced hypoglycae- ate the effects of drugs which accelerate [70],
mia [24]. Accordingly, the relationship of gastric potentiate the effects of drugs that delay, gastric
emptying with glycaemia is bidirectional (Fig. 3). emptying [71], and affect both gastro-­
Guidelines for measurement of gastric emptying pyloroduodenal motility [72] and gastric electri-
state that blood glucose concentrations should be cal activity, in health. Prostaglandins [73] and
<15 mmol/L at the time of the test while avoiding nitric oxide (NO) [74] have been suggested as
hypoglycaemia [43]. potential mediators of these effects. These stud-
Acute hyperglycaemia (16–20  mmol/L) was ies employed glucose insulin ‘clamps’ to induce
first reported to slow gastric emptying in 1962 hyperglycaemia rapidly. Aigner et  al. [75]
[68], although Stunkard and Wolff had observed recently evaluated the effects of spontaneous
in 1956 that intravenous glucose, presumably fluctuations in fasting glucose on gastric empty-
leading to hyperglycaemia, abolished the so-­ ing in type 1 diabetes and failed to observe a rela-
called gastric hunger contractions [69]. tionship, although the elevations in blood glucose
Hyperglycaemia was shown to have a variable, that they observed were, in most cases, modest.

More rapid gastric emptying increases


postprandial glycaemia

Gastric emptying Postprandial blood


glucose

Acute hyperglycaemia slows gastric


emptying, while hypoglycaemia
accelerates it − +

GLP-1 slows
More rapid gastric emptying Beta cell Alpha cell
gastric emptying (insulin) (glucagon)
increases GLP-1
and GIP secretion

GLP-1, GIP increase


insulin
GLP-1, GIP
GLP-1 suppresses
glucagon

Fig. 3  Schema of the inter-dependent relationships between gastric emptying, postprandial glycaemia, and incretin
hormones [67]
478 K. L. Jones et al.

Accordingly, further studies, to clarify the effect the robust acceleration of gastric emptying by
of hyperglycaemia on gastric emptying, are insulin-induced hypoglycaemia is likely to repre-
required, including evaluation of the effect of sent a hitherto, poorly appreciated, but important
treatment-induced, improved glycemic control counter-regulatory mechanism.
where current information is limited. There is
uncontrolled evidence that successful pancreas-­
kidney transplantation may reverse gastroparesis 4.4 Treatment
[76]; there is no information about the effect of
islet cell transplantation. The insights summarised in the preceding section
In contrast to the effect of hyperglycaemia, attest to the implicit importance of definition of
acute hypoglycaemia (blood glucose ~2.0 to the pathophysiology of a disease in order to
2.6 mmol/L) markedly accelerates gastric empty- develop approaches to effective management.
ing of solids and liquids in healthy subjects [77] Given that many of the significant advances are
and both complicated [77] and longstanding [78], recent, it is perhaps not surprising that, despite its
type 1 diabetes, the latter including patients with substantial health burden, few treatment options
cardiovascular autonomic dysfunction and, in for diabetic gastroparesis are available and their
some cases, gastroparesis (Fig.  4). The magni- efficacy is, in general, limited. The initial dogma
tude of this acceleration has been shown to be that making the stomach pump/empty ‘better’
dependent on the degree of hypoglycaemia [79], would result in symptom improvement, albeit
as well as vagal function [80] and, unlike the neu- intuitively illogical, has compromised progress
roendocrine responses to hypoglycaemia (e.g. [82]. Unfortunately, much of the evidence base is
stimulation of catecholamines), not attenuated by also derived from poorly designed studies and
a single episode of antecedent hypoglycaemia there is a lack of high quality randomised, con-
[81]. There is no information about the impact of trolled trials (Table 2).
insulin-induced hypoglycaemia on gastric emp- Management of gastroparesis, whether
tying in a large number of individuals with directed at symptom relief, improvement in gly-
insulin-­treated diabetes in whom the awareness caemic control and/or nutritional status should,
of hypoglycaemia is impaired. The more rapid like all therapies, ideally be individualised.
intestinal carbohydrate absorption resulting from General dietary recommendations, consumption

Solid Liquid
(100g minced beef) (150ml water)
100 100 Hypoglycaemia
Euglycaemia
80 p <0.01 80

Retention 60 60 p <0.05
(%)
40 40

20 20

0 0
0 15 30 45 60 75 90 105 120 0 15 30 45 60 75 90 105 120
Time (min) Time (min)

Fig. 4  Gastric emptying of solid (minced beef) and liquid from t  =  15–45  min) and euglycaemia in longstanding
(water) components of a meal, measured with scintigra- type 1 diabetes (n = 20). Hypoglycaemia accelerates gas-
phy, during hypoglycaemia (blood glucose ~2.6 mmol/L tric emptying [78]
Gastrointestinal Neuropathy 479

Table 2  Management of symptomatic diabetic prokinetic, was withdrawn from the market in
gastroparesis
2000 because of the potential for cardiac
• Optimise glycemic control arrhythmias.
  – In insulin-treated patients, address the timing of The dopamine (D2) antagonist, metoclo-
delivery of exogenous insulin to that of nutrient
absorption; consider continuous subcutaneous insulin pramide, can be administered subcutaneously,
delivery with concomitant continuous subcutaneous intranasally, or orally (subcutaneous metoclo-
glucose monitoring (with high- and low-glucose pramide has been used to manage attacks of vom-
alarms) iting on an outpatient basis). The intranasal route
• Address nutritional status
may be effective in women, but not men [87].
  – Modify the size, composition, and frequency of
meals The use of metoclopramide is associated with a
  – Consider jejunal feeding risk of QTc prolongation and tardive dyskinesia
• Address delayed gastric emptying with a trial of [88]. Because of the latter, in many countries,
prokinetic therapy including the USA, metoclopramide can cur-
• Address the dominant symptom(s) (e.g. nausea) with rently only be used for short-term treatment (e.g.
‘symptom-specific’ therapy
12  weeks). Domperidone is also a D2 receptor
• Address psychological co-morbidities
antagonist, but does not cross the blood–brain
barrier readily and is, accordingly, less prone to
of frequent, small meals and avoidance of food central nervous system adverse effects than meto-
that may provoke symptoms (particularly those clopramide [89]. Domperidone may also affect
high in fibre or fat), have, for the main part, not the QT interval and is not readily available in the
been evaluated rigorously [83, 84]. Moreover, USA.
there is anecdotal evidence that outcomes are The macrolide antibiotics, erythromycin and
only rarely satisfactory. In malnourished patients azithromycin, are motilin receptor agonists, fre-
in whom symptoms are refractory, a trial of naso-­ quently used ‘off-label’ for their prokinetic
jejunal feeding should certainly be considered. effects. Their long-term efficacy has not been
Modestly accelerated gastric emptying, as often established, tachyphylaxis to the acceleration of
observed in type 2 diabetes, does not appear to be emptying probably occurs, and there is also a risk
associated with symptoms. As discussed, drugs of QTc prolongation, particularly as the result of
which may slow gastric emptying should, ideally, drug interactions [90]. Mitemcinal, a motilin
be ceased, but this may not prove feasible. agonist, but without antibiotic properties, was
Optimising glycaemic control is logical and sup- shown to accelerate gastric emptying over
ported by uncontrolled data of the effects of sub- 3  months in diabetic gastroparesis but, unfortu-
cutaneous administration of insulin via a pump nately, failed to improve symptoms [36].
linked to continuous monitoring of subcutaneous Novel agents have been shown to accelerate
glucose concentrations [85]. Further studies are gastric emptying. One of these, relamorelin,
required. administered subcutaneously slowed efficacy for
As indicated, pharmacological treatment has symptom improvement, as well as accelerating
focussed on prokinetic medications designed to gastric emptying in phase 2 [91, 92]; phase 3 tri-
accelerate gastric emptying and, despite substan- als were, however, subsequently terminated. The
tial limitations, this remains the mainstay of cur- oral, highly selective 5HT-4 agonists, prucalo-
rent management. All of the prokinetic pride and velusetrag, were developed for the
medications have a significant adverse effect pro- management of constipation, but have been
file and with some, there is a reduction in effect shown to also accelerate gastric emptying.
with sustained administration (tachyphylaxis) Prucalopride has also been reported to accelerate
[86]. Not surprisingly, the relationship of symp- emptying in a small study of diabetic gastropare-
tomatic improvement with effects to accelerate sis [93]. Further studies are required. Recently,
gastric emptying is, at best, weak [84]. Cisapride, the selective 5HT-4 agonist, TAK-954, was
a widely used, and possibly the most effective, reported to normalise gastric emptying of a liquid
480 K. L. Jones et al.

meal in more critically ill patients with enteral that the frequency of vomiting was less when the
feeding intolerance than receiving metoclo- device was turned on [101]. Initial enthusiasm for
pramide [94]. Again, further studies are required. the potential use of intrapyloric botulinum toxin
Pharmacological therapy directed at specific injection was dampened by the negative outcome
symptoms may be useful, including antiemetics of sham-controlled trials [102, 103]. Gastric per-
such as 5HT-3 antagonists, phenothiazines, and oral endoscopic pyloromyotomy (G-POEM),
cannabinoids, none of which has been evaluated which is being evaluated as a treatment for gas-
adequately. The tachykinin receptor (neurokinin- troparesis, has not been evaluated in a controlled
1 receptor) antagonist, tradipitant, has been
­ trial. Acupuncture has been advocated for symp-
reported to improve nausea in patients with gas- tom relief in gastroparesis but, in a recent
troparesis, but the study was not powered ade- Cochrane review the major limitations of the evi-
quately to evaluate effects in the diabetic subgroup dence are apparent [104].
alone [95]. The tricyclic antidepressant, nortripty-
line, has been reported to be ineffective in symp-
tomatic, idiopathic gastroparesis [96]. Abell et al. 5 Gallbladder
[97] recently reported that sepiapterin, which
increases levels of NO, is associated with an The prevalence of cholelithiasis is increased
improvement in gastric accommodation in women markedly in type 2 diabetes, which is not surpris-
with diabetic gastroparesis, as evaluated by a ing given the high prevalence of well recognised
nutrient satiety test [98]. risk factors, including obesity and hypertriglyc-
Drugs used in the management of chronic eridaemia. Autonomic neuropathy (and possibly,
abdominal pain, such as gabapentin and pregaba- delayed gastric emptying) is also likely to be
lin, have not been evaluated formally in diabetic important in the pathogenesis of gallstones, sup-
gastroparesis. ported by observations that postprandial gallblad-
An improved understanding of the mecha- der emptying is frequently impaired [105] and
nisms underlying the gastric motor and sensory that acute hyperglycaemia inhibits gallbladder
dysfunctions in diabetes is likely to be central to emptying in health [106]. Interestingly, the use of
the development of novel, and more effective, GLP-1RAs, particularly liraglutide, is associated
pharmacotherapy. The design of future clinical with an increased risk of gallbladder/biliary dis-
trials should also focus on the most relevant end- ease for uncertain reasons [107], but possibly, in
points, which will influence trial design and sub- part, by slowing gallbladder contraction.
ject selection criteria. The US FDA has recently
recommended that trials evaluating novel treat-
ments for gastroparesis should have patient-­ 6 Small and Large Intestine
reported outcomes as the primary endpoint [99].
Invasive therapies for diabetic gastroparesis While diabetic enteropathy occurs frequently
include gastric electrical stimulation, intrapyloric and also represents an important cause of mor-
injections of botulinum toxin, and pyloromyot- bidity, there is much less information about it
omy, none of which has established efficacy. when compared to gastroparesis. This is despite
Gastric electrical stimulation (approved by the the fact that ‘diabetic diarrhoea’, attributed to
FDA on humanitarian grounds) involves the autonomic neuropathy [108], was first described
delivery of low energy, high frequency pulses to in 1936 [109], and the prevalence of symptoms
the antrum and does not accelerate gastric empty- of diarrhoea, constipation, and faecal inconti-
ing. The outcomes of initial ‘open-label’ studies nence is much higher in people with diabetes in
were promising, but not supported by subsequent, community studies [10]. In tertiary populations,
more rigorous, evaluation [100]. However, a the prevalence of symptoms is even greater
recent study of patients with refractory vomiting about 20% for diarrhoea and ~60% for constipa-
(with or without delayed gastric emptying) found tion [110].
Gastrointestinal Neuropathy 481

6.1 Diagnosis Table 3  Potential causes of diarrhoea in patients with


diabetes

In most cases, diabetic enteropathy represents a • Medications


diagnosis of exclusion. Medications used fre-   – Metformin
 – α-glucosidase inhibition—acarbose, voglibose, or
quently in the management of diabetes are par-
miglitol
ticularly metformin (which may cause diarrhoea   – GLP-1 receptor agonists
by inhibiting bile acid absorption),   – Orlistat
α-glucosidase inhibitors (acarbose, miglitol,   – Sorbitol, mannitol, or xylitol
voglibose), GLP-­1RAs and SGLT-2 inhibitors • Small intestinal bacterial overgrowth
(Table 3). Small intestinal bacterial overgrowth • Coeliac disease (especially in type 1 diabetes)
can be diagnosed by jejunal aspiration and cul- • Pancreatic exocrine insufficiency
ture, and/or breath tests, but both approaches • Microscopic colitis
have substantial limitations [111]. The preva- • Endocrine disorders (especially in type 1 diabetes)
  – Hyperthyroidism
lence of coeliac disease is increased substan-
  – Adrenal insufficiency
tially (~5%) in type 1 diabetes, so that
• ‘Diabetic diarrhoea’
serological screening is justified, even in the • Faecal incontinence
absence of symptoms. Diabetes, usually inap-
propriately regarded as type 2, is not infre-
Table 4  Medications associated with slowing of gastric
quently associated with pancreatic exocrine emptying
insufficiency (type 3c diabetes), where the
• Opiates
demonstration of a low level of faecal elastase
• GLP-1 receptor agonists
is diagnostically useful and pancreatic enzyme • Pramlintide
replacement has the potential to improve both • Anticholinergics
symptoms and glycaemic control, as well as • Levodopa
nutritional status. In patients with diarrhoea, • Metformin (?)
microscopic colitis, albeit rare and associated • Calcium channel antagonists
with autoimmune disorders, should be consid- • Beta-blockers
ered [112]. Hyperthyroidism and Addison’s • Somatostatin analogs (e.g. octreotide)
disease occur more frequently in type 1 diabe-
tes and may cause diarrhoea. Importantly, dis-
orders that frequently affect the general for slow transit, or a defecatory disorder, should
population, particularly irritable bowel syn- be considered.
drome, also occur frequently in people who
have diabetes. Diabetes has been reported to be
a risk factor for Clostridium difficile infection, 6.2 Pathophysiology
where metformin may be protective [113].
Approaches to the diagnosis of constipation Intestinal contractile activity may be increased,
and faecal incontinence in diabetes are similar to decreased, or both (as well as uncoordinated), as
those in individuals without diabetes. In patients a result of autonomic neuropathy in diabetes,
with constipation, medication should be reviewed which the inference is that transit may be ‘nor-
and those that frequently cause constipation ide- mal’, accelerated, or delayed. Gastroparesis has
ally ceased (e.g. opioids, anticholinergics, cal- been reported to be associated with abnormal
cium channel antagonists) (Table  4). For mild small intestinal motility in ~80% of cases [114].
constipation, lifestyle modifications, including Rodent models of diabetes have demonstrated
increases in physical exercise and dietary fibre, that adrenergic innervation may be impaired and
are usually recommended. A therapeutic trial of a contribute to diarrhoea, in part by modifying
laxative (bulk, osmotic, or stimulatory) is often fluid and electrolyte absorption [112]. Diabetic
indicated. If this proves ineffective, evaluation diarrhoea is associated with alterations in both
482 K. L. Jones et al.

gastrointestinal transit and intraluminal pH [115]. mosapride [124] and the cholinesterase inhibitor,
In health, acute elevations in blood glucose have pyridostigmine [125]. Lubiprostone, which acti-
been reported to affect small intestinal motility vates CIC-2 chloride channels, has been reported
[116], but the amount of information is limited. to accelerate colonic transit and increase bowel
In type 1 diabetes, small intestinal glucose frequency in diabetes [126]. There is also evi-
absorption is abnormally increased during hyper- dence that linaclotide, which increases guanylate
glycaemia for uncertain reasons [117]. As is the cyclase-C, may be useful [127], but further stud-
case with gastroparesis, the concept that vagal ies are required. In some cases, anorectal dys-
dysfunction is the dominant abnormality in dia- function responds to biofeedback therapy [128].
betic enteropathy has been broadened with the
recognition of abnormalities in the ICCs and
nNOS [54]. 7 Modulation of Gastric
Small intestinal bacterial overgrowth occurs Emptying and Small
frequently, albeit with variable estimates, and is Intestinal Motility to Improve
likely to be secondary to altered small intestinal Glycaemic Control
motility. While there is relatively little informa-
tion about colonic motility, it is clear that Gastric emptying is a major determinant of post-
colonic transit is frequently delayed, and consti- prandial glycaemia in health, as well as diabetes,
pation, more likely [118], in individuals with accounting for about a third of the variance in the
cardiac autonomic neuropathy [110]. In patients initial rise in blood glucose [129–131]. This is of
undergoing colectomy, a loss of myenteric, par- particular relevance to the achievement of good
ticularly inhibitory nitrergic neurons, has been glycaemic control in diabetes (often classified as
reported [119]. a glycated haemoglobin (HbA1c) level <7.0%),
The abnormalities in anorectal function, which is highly desirable to minimise the risk of
investigated most often in patients with faecal the development and progression of the micro-
incontinence, are heterogeneous with changes in vascular complications of diabetes, i.e. retinopa-
internal and external anal sphincter function and thy, nephropathy, and neuropathy, markedly.
impaired rectal sensation to distension [120, When HbA1c is >7.0% but <8.0%, the postpran-
121]. Acute hyperglycaemia may reduce anal dial rise, rather than preprandial (fasting), blood
sphincter pressures [122]. glucose, is the dominant determinant (even in
patients managed with insulin) [132]. In a 75  g
oral glucose tolerance test, it is not widely appre-
6.3 Treatment ciated that the 60 min plasma glucose (rather than
the fasting and 120  min glucose levels that are
As indicated, the diagnosis of diabetic enteropa- used diagnostically) is the strongest predictor of
thy is essentially one of exclusion and there is a the future development of type 2 diabetes [133]
lack of specific therapy. Small intestinal bacterial and this value is closely related to the rate of gas-
overgrowth should be treated with antibiotics tric emptying in subjects with impaired glucose
(most commonly rifaximin), but frequently tolerance or type 2 diabetes [131]. It is, accord-
relapses. When ‘diabetic diarrhoea’ is refractory, ingly, surprising that the importance of the rate of
oral clonidine (an α-2 agonist) and subcutaneous gastric emptying which, by definition, determines
octreotide may prove effective [112]. There is the delivery of carbohydrates to the small intes-
limited information about the treatment of consti- tine, has until recently received little attention, in
pation in diabetes. There is evidence, albeit lim- contrast to the focus on the relevance of the car-
ited, that flaxseed may be more effective than bohydrate content of a meal. Modulation of
psyllium and also improve glycaemic control in ­gastric emptying, by dietary or pharmacological
type 2 diabetes [123]. The outcomes of small means, has been shown to have the capacity to
studies support the efficacy of the 5HT-4 agonist, affect postprandial glycaemic excursions pro-
Gastrointestinal Neuropathy 483

foundly [40, 134, 135]. For example, in type 2 ment of gastric emptying, perhaps using a stable
patients, slowing of emptying by a nutrient ‘pre- isotope breath test, would allow more person-
load’ [134] or administration of morphine [40] alised, and hence, effective management. Studies
reduces the glycaemic response to a subsequent to explore this issue are indicated.
meal, while acceleration of emptying, as induced It should be appreciated that, in addition to the
by erythromycin [40], increases it significantly. rate of gastric emptying, small intestinal motility
Such observations have stimulated the develop- is a determinant of the glycaemic response to a
ment, and now widespread, use of drugs targeting meal [141], albeit very poorly characterised. In
slowing of gastric emptying to improve glycae- the majority of studies, the relative importance of
mic control in type 2 diabetes, particularly ‘short-­ changes in gastric emptying from those on small
acting’ GLP-1RAs (exenatide BID [136] and intestinal motility to postprandial glycaemia has
lixisenatide [41, 137]) and the amylin agonist, not been discriminated, in part because of the
pramlintide [138]. The magnitude of the slowing substantial technical challenges. The relevance of
of gastric emptying induced by these drugs is changes in small intestinal motility/transit has
dependent on the baseline rate of gastric empty- been established by studies in which carbohy-
ing [136] (so that patients with gastroparesis do drate has been infused directly into the small
not appear to be susceptible to further slowing) intestine [142]. It is also recognised that GLP-­
and predictive of the extent of the reduction in 1RAs may affect small intestinal motility [143].
postprandial glucose [41, 136] (Fig. 5). While it This is, again, an area where further research is
has been suggested that the more recently devel- indicated.
oped ‘long-acting’ GLP-1RAs (e.g. liraglutide,
exenatide QW) do not slow gastric emptying
with chronic administration, i.e. their glucose-­ 8 Relevance of Gastric
lowering capacity solely reflects insulinotropic Emptying and Small
and glucagonostatic properties [139], recent Intestinal Motility
studies have shown that this is not the case, i.e. to Postprandial Hypotension
there is a sustained slowing, albeit probably less
than that induced by short-acting GLP-1RAs [42, Postprandial hypotension, usually defined as a
140]. These considerations are of particular rele- fall in systolic blood pressure of >20  mmHg
vance to the frequent use of a combination of a within 2 h of a meal, has been estimated to occur
‘short’- or ‘long’-acting GLP-1RA with a basal in 20–40% of individuals with longstanding type
insulin in the management of type 2 diabetes. In 1 or type 2 diabetes; a prevalence which is prob-
this situation, it is possible that routine assess- ably greater than that of orthostatic hypotension,

Fig. 5 Relationship 60 Placebo


between the magnitude 5µg exenatide
Postprandial glucose AUC(0-3h)

of glucose lowering and 50 10µg exenatide


slowing of gastric
emptying in type 2
40 r = -0.49, P=0.0004
(mmol/L*h/L)

diabetes (n = 17) [136]


30

20

10

0
0 1 2 3 4 5 6
T50 (h)
484 K. L. Jones et al.

a Gastric emptying b Systolic blood pressure

100 Placebo 145 Placebo


Lixisenatide Lixisenatide
80
135
% Retention

60

mmHg
125
40

20 115
P <0.001 P < 0.001
0 105
0 30 60 90 120 150 180 BL 15 30 45 60 75 90 105 120
Time (min) Time (min)

Fig. 6  Acute effects of lixisenatide (10 μg sc) on (a). gas- patients with type 2 diabetes (n = 15). Lixisenatide mark-
tric emptying of a 75  g oral glucose load, measured by edly slows gastric emptying and attenuates the magnitude
scintigraphy and (b). postprandial blood pressure in of the postprandial fall in blood pressure [41]

which is much better characterised [144]. In water, attenuates the fall [150]. It has recently
addition to diabetes, postprandial hypotension been shown that acute administration of both
occurs frequently in a number of neurological exogenous (intravenous) GLP-1 [151] and the
disorders associated with autonomic dysfunc- short-acting GLP-1RA, lixisenatide [41] mark-
tion, such as Parkinson’s disease and is also edly attenuates the fall in systolic blood pressure
common in apparently ‘healthy’ older individu- induced by oral glucose in type 2 diabetes, prob-
als [145]. While there is, unfortunately, a lack of ably primarily by slowing gastric emptying.
intervention studies to support direct causality, While additional studies are required prior to
postprandial hypotension is strongly associated clinical translation, the outcomes are certainly
with an increased risk of a number of adverse promising and support the concept that, in many
sequelae, particularly syncope and falls [146] ways, postprandial hypotension should be
and it also appears to be an independent risk fac- regarded as a ‘gastrointestinal’, rather than ‘car-
tor for mortality [147]. In the broadest sense, diovascular’, disorder and that dietary and/or
postprandial hypotension can be regarded as a pharmacological strategies which slow gastric
manifestation of autonomic dysfunction in dia- emptying are likely to prove useful in manage-
betes—a substantial increase in sympathetic ment (Fig. 6).
activity is required to maintain blood pressure
postprandially [148] consequent to the postpran-
dial diversion of blood to increase splanchnic 9 Conclusions
blood flow. However, it is now appreciated that
‘gastrointestinal’ factors unrelated to increased Gastrointestinal function, even when normal, is
intestinal blood flow per se, specifically gastric now recognised to be central to the management
distension, gastric emptying and the release of of type 1 and type 2 diabetes. The outcomes of
gastrointestinal hormones, are important in the recent studies capitalising on the novel investiga-
pathogenesis of postprandial hypotension and tive techniques represent a major paradigm shift
have implications for strategies for effective with the recognition that disordered gastrointesti-
management, which is currently lacking [145]. nal autonomic function occurs frequently, that
In type 2 diabetes the magnitude of the fall in the abnormalities are heterogeneous and often
systolic blood pressure after an oral glucose load associated with clinically relevant impacts on
is greater when the rate of gastric emptying is gastrointestinal symptoms, glycaemic control
relatively more rapid [149], while an increase in and/or postprandial blood pressure. Therapeutic
gastric distension, such as induced by drinking development has been compromised, to some
Gastrointestinal Neuropathy 485

extent, by the pursuit of concepts which proved 11. Petrick JL, Li N, Anderson LA, Bernstein L, Corley
DA, El Serag HB, et  al. Diabetes in relation to
oversimplistic and, in retrospect, were somewhat Barrett’s esophagus and adenocarcinomas of the
naive. An improved characterisation of dysfunc- esophagus: a pooled study from the International
tions appears pivotal to the development of novel Barrett’s and Esophageal Adenocarcinoma
and more effective therapies. The latter must also Consortium. Cancer. 2019;125:4210–23.
12. Roman S, Marjoux S, Thivolet C, Mion
be evaluated in well-designed controlled trials, F. Oesophageal function assessed by high-resolution
which is, itself, challenging. manometry in patients with diabetes and inadequate
glycaemic control. Diabet Med. 2014;31:1452–9.
13. Kinekawa F, Kubo F, Matsuda K, Fujita Y, Tomita
T, Uchida Y, et al. Relationship between esophageal
References dysfunction and neuropathy in diabetic patients. Am
J Gastroenterol. 2001;96:2026–32.
1. Du YT, Rayner CK, Jones KL, Talley NJ, Horowitz 14. Boeckxstaens GE, Horowitz M, Bermingham H,
M.  Gastrointestinal symptoms in diabetes: preva- Holloway RH. Physiological variations in blood glu-
lence, assessment, pathogenesis, and management. cose concentration affect oesophageal motility and
Diabetes Care. 2018;41:627–37. sensation in normal subjects. Neurogastroenterol
2. Quan C, Talley NJ, Jones MP, Spies J, Horowitz Motil. 1997;9:239–46.
M.  Gain and loss of gastrointestinal symptoms in 15. Holloway RH, Tippett MD, Horowitz M, Maddox
diabetes mellitus: associations with psychiatric AF, Moten J, Russo A. Relationship between esoph-
disease, glycemic control, and autonomic neuropa- ageal motility and transit in patients with type I dia-
thy over 2 years of follow-up. Am J Gastroenterol. betes mellitus. Am J Gastroenterol. 1999;94:3150–7.
2008;103:2023–30. 16. Frokjaer JB, Brock C, Brun J, Simren M, Dimcevski
3. Quan C, Talley NJ, Cross S, Jones M, Hammer J, G, Funch-Jensen P, et  al. Esophageal distension
Giles N, et  al. Development and validation of the parameters as potential biomarkers of impaired
diabetes bowel symptom questionnaire. Aliment gastrointestinal function in diabetes patients.
Pharmacol Ther. 2003;17:1179–87. Neurogastroenterol Motil. 2012;24:1016–e544.
4. Revicki DA, Speck RM, Lavoie S, Puelles J, Kuo 17. Brock C, Softeland E, Gunterberg V, Frokjaer JB,
B, Camilleri M, et al. The American neurogastroen- Lelic D, Brock B, et al. Diabetic autonomic neuropa-
terology and motility society gastroparesis cardinal thy affects symptom generation and brain-gut axis.
symptom index-daily diary (ANMS GCSI-DD): Diabetes Care. 2013;36:3698–705.
psychometric evaluation in patients with idiopathic 18. Dumic I, Nordin T, Jecmenica M, Stojkovic Lalosevic
or diabetic gastroparesis. Neurogastroenterol Motil. M, Milosavljevic T, Milovanovic T. Gastrointestinal
2019;31:e13553. tract disorders in older age. Can J Gastroenterol
5. Rayner CK, Jones KL, Horowitz M. Semaglutide vs Hepatol. 2019;2019:6757524.
placebo as an adjunct to intensive behavioral therapy 19. Badillo R, Francis D.  Diagnosis and treatment of
and body weight in adults with overweight or obe- gastroesophageal reflux disease. World J Gastrointest
sity. JAMA. 2021;326:1213–4. Pharmacol Ther. 2014;5:105–12.
6. Horowitz M, Maddox AF, Wishart JM, Harding PE, 20. Kassander P. Asymptomatic gastric retention in dia-
Chatterton BE, Shearman DJ. Relationships between betics (gastroparesis diabeticorum). Ann Intern Med.
oesophageal transit and solid and liquid gastric 1958;48:797–812.
emptying in diabetes mellitus. Eur J Nucl Med. 21. Rundles RW.  Diabetic neuropathy. General review
1991;18:229–34. with report of 125 cases. Medicine. 1945;24:111–60.
7. Lluch I, Ascaso JF, Mora F, Minguez M, Pena A, 22. Watson LE, Xie C, Wang X, Li Z, Phillips LK, Sun
Hernandez A, et al. Gastroesophageal reflux in dia- Z, et  al. Gastric emptying in patients with well-­
betes mellitus. Am J Gastroenterol. 1999;94:919–24. controlled type 2 diabetes compared to non-diabetic
8. Horowitz M, Smout AJPM.  Disordered esopha- young and older controls. J Clin Endocrinol Metab.
geal and gastric motility in diabetes mellitus. Adv 2019;104:3311–9.
Endocrinol Metab. 1993;4:81–114. 23. Brener W, Hendrix TR, McHugh PR. Regulation of
9. Gokturk S, Akyuz F, Arici S, Alpaslan B, Ormeci A, the gastric emptying of glucose. Gastroenterology.
Soyer OM, et al. Gastroesophageal reflux in asymp- 1983;85:76–82.
tomatic patients with diabetes: an impedance study 24. Phillips LK, Deane AM, Jones KL, Rayner CK,
diabetes, obesity and gastroesophageal reflux. Exp Horowitz M.  Gastric emptying and glycaemia in
Clin Endocrinol Diabetes. 2020;128:52–8. health and diabetes mellitus. Nat Rev Endocrinol.
10. Bytzer P, Talley NJ, Leemon M, Young LJ, Jones 2015;11:112–28.
MP, Horowitz M.  Prevalence of gastrointestinal 25. Bharucha AE, Batey-Schaefer B, Cleary PA, Murray
symptoms associated with diabetes mellitus: a JA, Cowie C, Lorenzi G, et al. Delayed gastric empty-
population-­based survey of 15,000 adults. Arch ing is associated with early and long-term hypergly-
Intern Med. 2001;161:1989–96.
486 K. L. Jones et al.

cemia in type 1 diabetes mellitus. Gastroenterology. diabetes management. Neurogastroenterol Motil.


2015;149:330–9. 2006;18:405–7.
26. Chang J, Russo A, Bound M, Rayner CK, Jones 40. Gonlachanvit S, Hsu CW, Boden GH, Knight LC,
KL, Horowitz M.  A 25-year longitudinal evalua- Maurer AH, Fisher RS, et al. Effect of altering gas-
tion of gastric emptying in diabetes. Diabetes Care. tric emptying on postprandial plasma glucose con-
2012;35:2594–6. centrations following a physiologic meal in type-II
27. Wang YR, Fisher RS, Parkman HP.  Gastroparesis-­ diabetic patients. Dig Dis Sci. 2003;48:488–97.
related hospitalizations in the United States: trends, 41. Jones KL, Rigda RS, Buttfield MDM, Hatzinikolas
characteristics, and outcomes, 1995-2004. Am J S, Pham HT, Marathe CS, et al. Effects of lixisenatide
Gastroenterol. 2008;103:313–22. on postprandial blood pressure, gastric emptying and
28. Perano SJ, Rayner CK, Kritas S, Horowitz M, glycaemia in healthy people and people with type 2
Donaghue K, Mpundu-Kaambwa C, et  al. Gastric diabetes. Diabetes Obes Metab. 2019;21:1158–67.
emptying Is more rapid in adolescents with type 1 42. Jones KL, Huynh LQ, Hatzinikolas S, Rigda RS,
diabetes and impacts on postprandial glycemia. J Phillips LK, Pham HT, et al. Exenatide once weekly
Clin Endocrinol Metab. 2015;100:2248–53. slows gastric emptying of solids and liquids in
29. Phillips WT, Schwartz JG, McMahan CA.  Rapid healthy, overweight people at steady-state concen-
gastric emptying of an oral glucose solu- trations. Diabetes Obes Metab. 2020;22:788–97.
tion in type 2 diabetic patients. J Nucl Med. 43. Abell TL, Camilleri M, Donohoe K, Hasler WL, Lin
1992;33:1496–500. HC, Maurer AH, et al. Consensus recommendations
30. Schwartz JG, McMahan CA, Green GM, Phillips for gastric emptying scintigraphy: a joint report of
WT.  Gastric emptying in Mexican Americans the American Neurogastroenterology and Motility
compared to non-Hispanic whites. Dig Dis Sci. Society and the Society of Nuclear Medicine. J Nucl
1995;40:624–30. Med Technol. 2008;36:44–54.
31. Wang X, Xie C, Marathe CS, Malbert CH, Horowitz 44. Borg MJ, Bound M, Grivell J, Sun Z, Jones KL,
M, Jones KL, et al. Disparities in gastric emptying Horowitz M, et al. Comparative effects of proximal
and postprandial glycaemia between Han Chinese and distal small intestinal administration of metfor-
and Caucasians with type 2 diabetes. Diabetes Res min on plasma glucose and glucagon-like peptide-1,
Clin Pract. 2020;159:107951. and gastric emptying after oral glucose, in type 2
32. Jones KL, Horowitz M, Wishart JM, Maddox AF, diabetes. Diabetes Obes Metab. 2019;21:640–7.
Harding PE, Chatterton BE. Relationships between 45. Collins PJ, Horowitz M, Cook DJ, Harding PE,
gastric emptying, intragastric meal distribution and Shearman DJ.  Gastric emptying in normal sub-
blood glucose concentrations in diabetes mellitus. J jects--a reproducible technique using a single
Nucl Med. 1995;36:2220–8. scintillation camera and computer system. Gut.
33. Kuwelker S, Prichard DO, Bailey K, Bharucha 1983;24:1117–25.
AE. Relationship between symptoms during a gastric 46. Lee JS, Camilleri M, Zinsmeister AR, Burton DD,
emptying study, daily symptoms and quality of life in Kost LJ, Klein PD.  A valid, accurate, office based
patients with diabetes mellitus. Neurogastroenterol non-radioactive test for gastric emptying of solids.
Motil. 2021;33:e14154. Gut. 2000;46:768–73.
34. Jones KL, Russo A, Stevens JE, Wishart JM, Berry 47. Viramontes BE, Kim DY, Camilleri M, Lee JS,
MK, Horowitz M. Predictors of delayed gastric emp- Stephens D, Burton DD, et al. Validation of a stable
tying in diabetes. Diabetes Care. 2001;24:1264–9. isotope gastric emptying test for normal, accelerated
35. Parkman HP, Yates K, Hasler WL, Nguyen L, or delayed gastric emptying. Neurogastroenterol
Pasricha PJ, Snape WJ, et al. Similarities and differ- Motil. 2001;13:567–74.
ences between diabetic and idiopathic gastroparesis. 48. Sanaka M, Nakada K, Nosaka C, Kuyama Y.  The
Clin Gastroenterol Hepatol. 2011;9:1056–64; quiz Wagner-Nelson method makes the [13C]-breath test
e133–4. comparable to radioscintigraphy in measuring gastric
36. Stevens JE, Jones KL, Rayner CK, Horowitz emptying of a solid/liquid mixed meal in humans.
M. Pathophysiology and pharmacotherapy of gastro- Clin Exp Pharmacol Physiol. 2007;34:641–4.
paresis: current and future perspectives. Expert Opin 49. Saad RJ, Hasler WL. A technical review and clini-
Pharmacother. 2013;14:1171–86. cal assessment of the wireless motility capsule.
37. Ishii M, Nakamura T, Kasai F, Onuma T, Baba T, Gastroenterol Hepatol. 2011;7:795–804.
Takebe K. Altered postprandial insulin requirement 50. Hasler WL, May KP, Wilson LA, Van Natta M,
in IDDM patients with gastroparesis. Diabetes Care. Parkman HP, Pasricha PJ, et  al. Relating gastric
1994;17:901–3. scintigraphy and symptoms to motility capsule tran-
38. Lysy J, Israeli E, Strauss-Liviatan N, Goldin sit and pressure findings in suspected gastroparesis.
E. Relationships between hypoglycaemia and gastric Neurogastroenterol Motil. 2018;30:e13196.
emptying abnormalities in insulin-treated diabetic 51. Bartholome R, Salden B, Vrolijk MF, Troost FJ,
patients. Neurogastroenterol Motil. 2006;18:433–40. Masclee A, Bast A, et al. Paracetamol as a post pran-
39. Horowitz M, Jones KL, Rayner CK, Read NW. dial marker for gastric emptying, A food-drug inter-
‘Gastric’ hypoglycaemia--an important concept in action on absorption. PLoS One. 2015;10:e0136618.
Gastrointestinal Neuropathy 487

52. Horowitz M, Rayner CK, Marathe CS, Wu T, Jones 66. Schvarcz E, Palmer M, Aman J, Lindkvist B,
KL.  Glucagon-like peptide-1 receptor agonists and Beckman KW. Hypoglycaemia increases the gastric
the appropriate measurement of gastric emptying. emptying rate in patients with type 1 diabetes mel-
Diabetes Obes Metab. 2020;22:2504–6. litus. Diabet Med. 1993;10:660–3.
53. Grover M, Bernard CE, Pasricha PJ, Lurken 67. Marathe CS, Rayner CK, Jones KL, Horowitz
MS, Faussone-Pellegrini MS, Smyrk TC, et  al. M.  Relationships between gastric emptying, post-
Clinical-histological associations in gastropa- prandial glycemia, and incretin hormones. Diabetes
resis: results from the Gastroparesis Clinical Care. 2013;36:1396–405.
Research Consortium. Neurogastroenterol Motil. 68. Aylett P. Gastric emptying and change of blood glu-
2012;24(531-9):e249. cose level, as affected by glucagon and insulin. Clin
54. Kashyap P, Farrugia G. Diabetic gastroparesis: what Sci. 1962;22:171–8.
we have learned and had to unlearn in the past 5 69. Stunkard AJ, Wolff HG. Studies on the physiology
years. Gut. 2010;59:1716–26. of hunger. I. The effect of intravenous administration
55. Yarandi SS, Srinivasan S.  Diabetic gastrointesti- of glucose on gastric hunger contractions in man. J
nal motility disorders and the role of enteric ner- Clin Invest. 1956;35:954–63.
vous system: current status and future directions. 70. Jones KL, Berry M, Kong MF, Kwiatek MA,
Neurogastroenterol Motil. 2014;26:611–24. Samsom M, Horowitz M. Hyperglycemia attenuates
56. Thazhath SS, Jones KL, Horowitz M, Rayner the gastrokinetic effect of erythromycin and affects
CK.  Diabetic gastroparesis: recent insights into the perception of postprandial hunger in normal sub-
pathophysiology and implications for man- jects. Diabetes Care. 1999;22:339–44.
agement. Expert Rev Gastroenterol Hepatol. 71. Plummer MP, Jones KL, Annink CE, Cousins CE,
2013;7:127–39. Meier JJ, Chapman MJ, et  al. Glucagon-like pep-
57. Chakraborty S, Halland M, Burton D, Desai A, Neja tide 1 attenuates the acceleration of gastric empty-
B, Low P, et al. GI dysfunctions in diabetic gastro- ing induced by hypoglycemia in healthy subjects.
enteropathy, their relationships with symptoms, and Diabetes Care. 2014;37:1509–15.
effects of a GLP-1 antagonist. J Clin Endocrinol 72. Rayner CK, Su YC, Doran SM, Jones KL, Malbert
Metab. 2019;104:1967–77. CH, Horowitz M.  The stimulation of antral motil-
58. Faussone-Pellegrini MS, Grover M, Pasricha ity by erythromycin is attenuated by hyperglycemia.
PJ, Bernard CE, Lurken MS, Smyrk TC, et  al. Am J Gastroenterol. 2000;95:2233–41.
Ultrastructural differences between diabetic 73. Hasler WL, Soudah HC, Dulai G, Owyang
and idiopathic gastroparesis. J Cell Mol Med. C. Mediation of hyperglycemia-evoked gastric slow-­
2012;16:1573–81. wave dysrhythmias by endogenous prostaglandins.
59. Harberson J, Thomas RM, Harbison SP, Parkman Gastroenterology. 1995;108:727–36.
HP.  Gastric neuromuscular pathology in gastropa- 74. Kuo P, Gentilcore D, Nair N, Stevens JE, Wishart
resis: analysis of full-thickness antral biopsies. Dig JM, Lange K, et al. The nitric oxide synthase inhibi-
Dis Sci. 2010;55:359–70. tor, Ng-nitro-L-arginine-methyl-ester, attenuates the
60. Camilleri M, Chedid V, Ford AC, Haruma K, delay in gastric emptying induced by hyperglycae-
Horowitz M, Jones KL, et al. Gastroparesis. Nat Rev mia in healthy humans. Neurogastroenterol Motil.
Dis Primers. 2018;4:41. 2009;21:1175–e103.
61. Kashyap P, Farrugia G.  Oxidative stress: key 75. Aigner L, Becker B, Gerken S, Quast DR, Meier
player in gastrointestinal complications of diabetes. JJ, Nauck MA.  Day-to-day variations in fasting
Neurogastroenterol Motil. 2011;23:111–4. plasma glucose do not influence gastric emptying
62. Mazzone A, Bernard CE, Strege PR, Beyder A, in subjects with type 1 diabetes. Diabetes Care.
Galietta LJ, Pasricha PJ, et  al. Altered expression 2021;44:479–88.
of Ano1 variants in human diabetic gastroparesis. J 76. Gaber AO, Oxley D, Karas J, Cardoso S, Hathaway
Biol Chem. 2011;286:13393–403. D, Shokouh-Amiri MH, et  al. Changes in gas-
63. Puthanmadhom Narayanan S, O’Brien D, Sharma tric emptying in recipients of successful com-
M, Miller K, Adams P, Passos JF, et  al. Duodenal bined pancreas-­ kidney transplants. Dig Dis.
mucosal mitochondrial gene expression is associated 1991;9:437–43.
with delayed gastric emptying in diabetic gastroen- 77. Schvarcz E, Palmer M, Aman J, Berne
teropathy. JCI. Insight. 2021;6:e143596. C. Hypoglycemia increases the gastric emptying rate
64. Schvarcz E, Palmer M, Aman J, Horowitz M, in healthy subjects. Diabetes Care. 1995;18:674–6.
Stridsberg M, Berne C.  Physiological hyperglyce- 78. Russo A, Stevens JE, Chen R, Gentilcore D, Burnet
mia slows gastric emptying in normal subjects and R, Horowitz M, et al. Insulin-induced hypoglycemia
patients with insulin-dependent diabetes mellitus. accelerates gastric emptying of solids and liquids
Gastroenterology. 1997;113:60–6. in long-standing type 1 diabetes. J Clin Endocrinol
65. Fraser RJ, Horowitz M, Maddox AF, Harding PE, Metab. 2005;90:4489–95.
Chatterton BE, Dent J. Hyperglycaemia slows gas- 79. Murthy TA, Grivell J, Hatzinikolas S, Chapple LS,
tric emptying in type 1 (insulin-dependent) diabetes Chapman MJ, Stevens JE, et  al. Acceleration of
mellitus. Diabetologia. 1990;33:675–80. gastric emptying by insulin-induced hypoglycemia
488 K. L. Jones et al.

is dependent on the degree of hypoglycemia. J Clin domized placebo-controlled crossover pilot trial.
Endocrinol Metab. 2021;106:364–71. Neurogastroenterol Motil. 2021;33:e13958.
80. Hjelland IE, Oveland NP, Leversen K, Berstad A, 94. Chapman MJ, Jones KL, Almansa C, Barnes CN,
Hausken T.  Insulin-induced hypoglycemia stimu- Nguyen D, Deane AM.  Blinded, double-dummy,
lates gastric vagal activity and motor function with- parallel-group, phase 2a randomized clinical trial
out increasing cardiac vagal activity. Digestion. to evaluate the efficacy and safety of a highly
2005;72:43–8. selective 5-Hydroxytryptamine Type 4 receptor
81. Kar P, Jones KL, Plummer MP, Ali Abdelhamid Y, agonist in critically ill patients with enteral feed-
Giersch EJ, Summers MJ, et  al. Antecedent hypo- ing intolerance. JPEN J Parenter Enteral Nutr.
glycemia does not attenuate the acceleration of gas- 2021;45:115–24.
tric emptying by hypoglycemia. J Clin Endocrinol 95. Carlin JL, Lieberman VR, Dahal A, Keefe MS, Xiao
Metab. 2017;102:3953–60. C, Birznieks G, et al. Efficacy and safety of tradipi-
82. Rayner CK, Jones KL, Horowitz M. Is making the tant in patients with diabetic and idiopathic gastro-
stomach pump better the answer to gastroparesis? paresis in a randomized, placebo-controlled trial.
Gastroenterology. 2019;156:1555–7. Gastroenterology. 2021;160(76-87):e4.
83. Olausson EA, Storsrud S, Grundin H, Isaksson 96. Parkman HP, Van Natta ML, Abell TL, McCallum
M, Attvall S, Simren M.  A small particle size diet RW, Sarosiek I, Nguyen L, et  al. Effect of nor-
reduces upper gastrointestinal symptoms in patients triptyline on symptoms of idiopathic gastropare-
with diabetic gastroparesis: a randomized controlled sis: the NORIG randomized clinical trial. JAMA.
trial. Am J Gastroenterol. 2014;109:375–85. 2013;310:2640–9.
84. Jalleh R, Marathe CS, Rayner CK, Jones KL, 97. Abell TL, Garcia LM, Wiener GJ, Wo JM, Bulat
Horowitz M.  Diabetic gastroparesis and glycaemic RS, Smith N. Effect of oral CNSA-001 (sepiapterin,
control. Curr Diab Rep. 2019;19:153. PTC923) on gastric accommodation in women
85. Koch KL, Hasler WL, Van Natta M, Calles-Escandon with diabetic gastroparesis: a randomized, placebo-­
J, Grover M, Pasricha PJ, et al. Satiety testing in dia- controlled, phase 2 trial. J Diabetes Complicat.
betic gastroparesis: effects of insulin pump therapy 2021;35:107961.
with continuous glucose monitoring on upper gas- 98. Jalleh RJ, Marathe CS, Jones KL, Horowitz
trointestinal symptoms and gastric myoelectrical M, Rayner CK.  Digesting the pathogenesis of
activity. Neurogastroenterol Motil. 2020;32:e13720. diabetic gastroparesis. J Diabetes Complicat.
86. Tesfaye S, Boulton AJ, Dyck PJ, Freeman R, 2021;35:107992.
Horowitz M, Kempler P, et  al. Diabetic neuropa- 99. Pasricha PJ, Camilleri M, Hasler WL, Parkman
thies: update on definitions, diagnostic criteria, esti- HP.  White paper AGA: gastroparesis: clinical
mation of severity, and treatments. Diabetes Care. and regulatory insights for clinical trials. Clin
2010;33:2285–93. Gastroenterol Hepatol. 2017;15:1184–90.
87. Parkman HP, Carlson MR, Gonyer 100. McCallum RW, Snape W, Brody F, Wo J, Parkman
D. Metoclopramide nasal spray is effective in symp- HP, Nowak T.  Gastric electrical stimulation with
toms of gastroparesis in diabetics compared to Enterra therapy improves symptoms from dia-
conventional oral tablet. Neurogastroenterol Motil. betic gastroparesis in a prospective study. Clin
2014;26:521–8. Gastroenterol Hepatol. 2010;8:947–54; quiz e116.
88. Lee A, Kuo B.  Metoclopramide in the treatment 101. Ducrotte P, Coffin B, Bonaz B, Fontaine S, Bruley
of diabetic gastroparesis. Expert Rev Endocrinol Des Varannes S, Zerbib F, et  al. Gastric electri-
Metab. 2010;5:653–62. cal stimulation reduces refractory vomiting in
89. Tornblom H.  Treatment of gastrointestinal auto- a randomized crossover trial. Gastroenterology.
nomic neuropathy. Diabetologia. 2016;59:409–13. 2020;158:506–14.e2.
90. Rayner CK, Horowitz M.  New management 102. Arts J, Holvoet L, Caenepeel P, Bisschops R, Sifrim
approaches for gastroparesis. Nat Clin Pract D, Verbeke K, et  al. Clinical trial: a randomized-­
Gastroenterol Hepatol. 2005;2:454–62; quiz 93. controlled crossover study of intrapyloric injec-
91. Lembo A, Camilleri M, McCallum R, Sastre R, tion of botulinum toxin in gastroparesis. Aliment
Breton C, Spence S, et al. Relamorelin reduces vom- Pharmacol Ther. 2007;26:1251–8.
iting frequency and severity and accelerates gas- 103. Friedenberg FK, Palit A, Parkman HP, Hanlon A,
tric emptying in adults with diabetic gastroparesis. Nelson DB.  Botulinum toxin A for the treatment
Gastroenterology. 2016;151(87-96):e6. of delayed gastric emptying. Am J Gastroenterol.
92. Camilleri M, McCallum RW, Tack J, Spence SC, 2008;103:416–23.
Gottesdiener K, Fiedorek FT.  Efficacy and safety 104. Kim KH, Lee MS, Choi TY, Kim TH. Acupuncture
of relamorelin in diabetics with symptoms of gas- for symptomatic gastroparesis. Cochrane Database
troparesis: a randomized, placebo-controlled study. Syst Rev. 2018;12:CD009676.
Gastroenterology. 2017;153:1240–50.e2. 105. Pazzi P, Scagliarini R, Gamberini S, Pezzoli
93. Andrews CN, Woo M, Buresi M, Curley M, Gupta A.  Review article: gall-bladder motor function
M, Tack J, et  al. Prucalopride in diabetic and con- in diabetes mellitus. Aliment Pharmacol Ther.
nective tissue disease-related gastroparesis: ran- 2000;14(Suppl 2):62–5.
Gastrointestinal Neuropathy 489

106. Gielkens HAJ, Van Oostayen JA, Frölich M, Biemond internal-anal-sphincter dysfunction. N Engl J Med.
I, Lamers CBHW, Masclee AAM.  Dose-dependent 1982;307:1666–71.
inhibition of postprandial gallbladder motility and 121. Sun WM, Katsinelos P, Horowitz M, Read
plasma hormone secretion during acute hyperglyce- NW.  Disturbances in anorectal function in patients
mia. Scand J Gastroenterol. 1998;33:1074–9. with diabetes mellitus and faecal incontinence. Eur J
107. Nauck MA, Muus Ghorbani ML, Kreiner E, Gastroenterol Hepatol. 1996;8:1007–12.
Saevereid HA, Buse JB, Investigators LP, Cobot 122. Russo A, Sun WM, Sattawatthamrong Y, Fraser R,
LT.  Effects of liraglutide compared with placebo Horowitz M, Andrews JM, et al. Acute hyperglyce-
on events of acute gallbladder or biliary disease in mia affects anorectal motor and sensory function in
patients with type 2 diabetes at high risk for cardio- normal subjects. Gut. 1997;41:494–9.
vascular events in the LEADER randomized trial. 123. Soltanian N, Janghorbani M.  Effect of flaxseed or
Diabetes Care. 2019;42:1912–20. psyllium vs. placebo on management of constipa-
108. Dooley CP, el Newihi HM, Zeidler A, Valenzuela tion, weight, glycemia, and lipids: a randomized
JE.  Abnormalities of the migrating motor complex trial in constipated patients with type 2 diabetes.
in diabetics with autonomic neuropathy and diar- Clin Nutr ESPEN. 2019;29:41–8.
rhea. Scand J Gastroenterol. 1988;23:217–23. 124. Ueno N, Inui A, Satoh Y.  The effect of mosapride
109. Bargen JA, Bollman JL, Kepler EJ. The “diarrhea of citrate on constipation in patients with diabetes.
diabetes” and steatorrhea of pancreatic insufficiency. Diabetes Res Clin Pract. 2010;87:27–32.
Mayo Clin Proc. 1936;11:737–42. 125. Bharucha AE, Low P, Camilleri M, Veil E, Burton D,
110. Phillips LK, Rayner CK, Jones KL, Horowitz M. An Kudva Y, et al. A randomised controlled study of the
update on autonomic neuropathy affecting the gas- effect of cholinesterase inhibition on colon function
trointestinal tract. Curr Diab Rep. 2006;6:417–23. in patients with diabetes mellitus and constipation.
111. Quigley EMM, Murray JA, Pimentel M.  AGA Gut. 2013;62:708–15.
Clinical practice update on small intestinal bacte- 126. Christie J, Shroff S, Shahnavaz N, Carter LA,
rial overgrowth: expert Review. Gastroenterology. Harrison MS, Dietz-Lindo KA, et al. A randomized,
2020;159:1526–32. double-blind, placebo-controlled trial to examine the
112. Selby A, Reichenbach ZW, Piech G, Friedenberg effectiveness of lubiprostone on constipation symp-
FK.  Pathophysiology, differential diagnosis, toms and colon transit time in diabetic patients. Am
and treatment of diabetic diarrhea. Dig Dis Sci. J Gastroenterol. 2017;112:356–64.
2019;64:3385–93. 127. Yang J, Lei Y. Comparison of the efficacy and safety
113. Eliakim-Raz N, Fishman G, Yahav D, Goldberg E, of different doses of linaclotide for patients with
Stein GY, Zvi HB, et al. Predicting Clostridium diffi- chronic constipation: a meta-analysis and Bayesian
cile infection in diabetic patients and the effect of met- analysis. Evid Based Complement Alternat Med.
formin therapy: a retrospective, case-­control study. 2021;2021:9923879.
Eur J Clin Microbiol Infect Dis. 2015;34:1201–5. 128. Wald A, Tunuguntla AK.  Anorectal sensorimotor
114. Camilleri M, Malagelada JR.  Abnormal intestinal dysfunction in fecal incontinence and diabetes mel-
motility in diabetics with the gastroparesis syn- litus. Modification with biofeedback therapy. N Engl
drome. Eur J Clin Investig. 1984;14:420–7. J Med. 1984;310:1282–7.
115. Sangnes DA, Dimcevski G, Frey J, Softeland 129. Horowitz M, Edelbroek MA, Wishart JM, Straathof
E.  Diabetic diarrhoea: a study on gastrointestinal JW.  Relationship between oral glucose tolerance
motility, pH levels and autonomic function. J Intern and gastric emptying in normal healthy subjects.
Med. 2021;290:1206. Diabetologia. 1993;36:857–62.
116. Russo A, Fraser R, Horowitz M. The effect of acute 130. Jones KL, Horowitz M, Carney BI, Wishart JM,
hyperglycaemia on small intestinal motility in nor- Guha S, Green L.  Gastric emptying in “early”
mal subjects. Diabetologia. 1996;39:984–9. noninsulin-dependent diabetes mellitus relationship
117. Rayner CK, Samsom M, Jones KL, Horowitz to oral glucose tolerance and appetite. J Nucl Med.
M. Relationships of upper gastrointestinal motor and 1996;37:1643–8.
sensory function with glycemic control. Diabetes 131. Marathe CS, Horowitz M, Trahair LG, Wishart JM,
Care. 2001;24:371–81. Bound M, Lange K, et al. Relationships of early and
118. Prasad VG, Abraham P. Management of chronic con- late glycemic responses with gastric emptying dur-
stipation in patients with diabetes mellitus. Indian J ing an oral glucose tolerance test. J Clin Endocrinol
Gastroenterol. 2017;36:11–22. Metab. 2015;100:3565–71.
119. Chandrasekharan B, Anitha M, Blatt R, Shahnavaz 132. Riddle M, Umpierrez G, DiGenio A, Zhou R,
N, Kooby D, Staley C, et  al. Colonic motor dys- Rosenstock J. Contributions of basal and postpran-
function in human diabetes is associated with dial hyperglycemia over a wide range of A1C levels
enteric neuronal loss and increased oxidative stress. before and after treatment intensification in type 2
Neurogastroenterol Motil. 2011;23(131-8):e26. diabetes. Diabetes Care. 2011;34:2508–14.
120. Schiller LR, Santa Ana CA, Schmulen AC, Hendler 133. Bergman M.  The 1-hour plasma glucose: common
RS, Harford WV, Fordtran JS. Pathogenesis of fecal link across the glycemic spectrum. Front Endocrinol
incontinence in diabetes mellitus: evidence for (Lausanne). 2021;12:752329.
490 K. L. Jones et al.

134. Ma J, Stevens JE, Cukier K, Maddox AF, Wishart to intraduodenal glucose infusion. Am J Physiol
JM, Jones KL, et al. Effects of a protein preload on Gastrointest Liver Physiol. 2010;299:G1326–33.
gastric emptying, glycemia, and gut hormones after 143. Thazhath SS, Marathe C, Wu T, Chang J, Khoo J,
a carbohydrate meal in diet-controlled type 2 diabe- Kuo P, et al. Effects of the glucagon-like peptide-1
tes. Diabetes Care. 2009;32:1600–2. (GLP-1) receptor agonist, exenatide, on small intes-
135. Kamruzzaman M, Horowitz M, Jones KL, Marathe tinal motility, flow and glucose absorption in healthy
CS.  Gut-based strategies to reduce postprandial subjects and type 2 diabetes. United European
glycaemia in type 2 diabetes. Front Endocrinol Gastroenterol J. 2014;2:A29.
(Lausanne). 2021;12:661877. 144. Mathias CJ, Jones KL. Postprandial hypotension in
136. Linnebjerg H, Park S, Kothare PA, Trautmann autonomic disorders. In: Mathias CJ, Bannister R,
ME, Mace K, Fineman M, et  al. Effect of exena- editors. Autonomic failure: a textbook of clinical
tide on gastric emptying and relationship to post- disorders of the autonomic nervous system. Oxford:
prandial glycemia in type 2 diabetes. Regul Pept. Oxford University Press; 2013. p. 354–70.
2008;151:123–9. 145. Trahair LG, Horowitz M, Jones KL.  Postprandial
137. Lorenz M, Pfeiffer C, Steinstrasser A, Becker RH, hypotension: a systematic review. J Am Med Dir
Rutten H, Ruus P, et  al. Effects of lixisenatide Assoc. 2014;15:394–409.
once daily on gastric emptying in type 2 diabetes- 146. Puisieux F, Bulckaen H, Fauchais AL, Drumez S,
-relationship to postprandial glycemia. Regul Pept. Salomez-Granier F, Dewailly P.  Ambulatory blood
2013;185:1–8. pressure monitoring and postprandial hypotension in
138. Samsom M, Szarka LA, Camilleri M, Vella A, elderly persons with falls or syncopes. J Gerontol A
Zinsmeister AR, Rizza RA. Pramlintide, an amylin Biol Sci Med Sci. 2000;55A:M535–M40.
analog, selectively delays gastric emptying: potential 147. Fisher AA, Davis MW, Srikusalanukul W, Budge
role of vagal inhibition. Am J Physiol Gastrointest MM.  Postprandial hypotension predicts all-cause
Liver Physiol. 2000;278:G946–51. mortality in older, low-level care residents. J Am
139. Drucker DJ, Buse JB, Taylor K, Kendall DM, Geriatr Soc. 2005;53:1313–20.
Trautmann M, Zhuang D, et  al. Exenatide once 148. Masuda Y, Kawamura A. Role of the autonomic ner-
weekly versus twice daily for the treatment of type vous system in postprandial hypotension in elderly
2 diabetes: a randomised, open-label, non-inferiority persons. J Cardiovasc Pharmacol. 2003;42(Suppl
study. Lancet. 2008;372:1240–50. 1):S23–6.
140. Halawi H, Khemani D, Eckert D, O’Neill J, Kadouh 149. Jones KL, Tonkin A, Horowitz M, Wishart JM,
H, Grothe K, et al. Effects of liraglutide on weight, Carney BI, Guha S, et  al. Rate of gastric empty-
satiation, and gastric functions in obesity: a ran- ing is a determinant of postprandial hypotension in
domised, placebo-controlled pilot trial. Lancet non-insulin-dependent diabetes mellitus. Clin Sci
Gastroenterol Hepatol. 2017;2:890–9. (Lond). 1998;94:65–70.
141. Thazhath SS, Marathe CS, Wu T, Chang J, Khoo 150. Shannon JR, Diedrich A, Biaggioni I, Tank J,
J, Kuo P, et al. The glucagon-like peptide 1 recep- Robertson RM, Robertson D, et  al. Water drinking
tor agonist exenatide inhibits small intestinal as a treatment for orthostatic syndromes. Am J Med.
motility, flow, transit, and absorption of glucose 2002;112:355–60.
in healthy subjects and patients with type 2 dia- 151. Trahair LG, Horowitz M, Stevens JE, Feinle-Bisset
betes: a randomized controlled trial. Diabetes. C, Standfield S, Piscitelli D, et al. Effects of exog-
2016;65:269–75. enous glucagon-like peptide-1 on blood pressure,
142. Kuo P, Bellon M, Wishart J, Smout AJ, Holloway heart rate, gastric emptying, mesenteric blood flow
RH, Fraser RJ, et al. Effects of metoclopramide on and glycaemic responses to oral glucose in older
duodenal motility and flow events, glucose absorp- individuals with normal glucose tolerance or type 2
tion, and incretin hormone release in response diabetes. Diabetologia. 2015;58:1769–78.
Diabetic Neuropathy: Clinical
Management—Genitourinary
Dysfunction in Diabetes

Vincenza Spallone, Enrico Finazzi Agrò,


Roberta Centello, Claudio Lecis, Luca Orecchia,
and Andrea M. Isidori

1 Introduction testing standardization, with consequent discrep-


ancies in epidemiological data, although some
Bladder and sexual dysfunction in diabetes advancement has been made mainly for erectile
include a variety of conditions dealing with two dysfunction (ED). Finally, they coexist in many
distinct functions. In addition to sharing changes patients pointing to common pathogenesis and an
in vascularization and innervation, however, they interrelationship at multiple levels including psy-
hold additional similarities. These are common chological aspects.
complications in people with diabetes reaching in
some manifestations a prevalence of over 50%,
the highest prevalence among diabetic complica- 2 Bladder Dysfunction
tions. They are burdened with a relevant impact in Diabetes
on quality of life (QoL) and on survival.
Notwithstanding, they are underdiagnosed and The traditional concept of diabetic cystopathy as
underestimated. For both bladder and sexual dys- the gradual development of decreased bladder
function, a shift has occurred from the attribution sensation, increased capacity, and impaired
of main pathogenetic responsibility to diabetic detrusor contractility and emptying [1] has given
neuropathy to a multiorgan and multifactorial way to the actual definition of diabetic bladder
pathophysiology. Their nature is recognized as dysfunction as a broad spectrum of abnormalities
multidimensional: organic, relational, and intra- and lower urinary tract symptoms (LUTS) [2].
psychic. However, both bladder and sexual dys- The watershed moment that changed the view of
function are still characterized by poor knowledge pathophysiology of diabetic cystopathy was the
and clarity in definition, in diagnostic criteria and observation that in patients with diabetes and
voiding symptoms the most frequent urodynamic
finding was detrusor instability (55%) and not
V. Spallone (*)
Endocrinology, Department of Systems Medicine, detrusor areflexia (10%) [3].
University of Rome Tor Vergata, Rome, Italy
e-mail: vincenza.spallone@uniroma2.it
E. Finazzi Agrò · L. Orecchia 2.1 Epidemiology of Diabetic
Urology, Department of Surgical Sciences, University Bladder Dysfunction
of Rome Tor Vergata, Rome, Italy
R. Centello · C. Lecis · A. M. Isidori In the general population urinary incontinence
Department of Experimental Medicine, Sapienza has been reported to occur in 5–69% of women
University of Rome, Rome, Italy

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 491
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7_28
492 V. Spallone et al.

and in 1–39% of men, while overactive bladder in ratio 2.56), possible diabetic polyneuropathy
8–42% of women and in 10–26% of men [4]. (odds ratio 1.97), and cardiovascular autonomic
Risk factors of urinary incontinence in women neuropathy (CAN) (odds ratio 2.07) [9, 10].
are pregnancy, vaginal delivery, pelvic organ pro- Interestingly, in 71% of cases LUTS and ED
lapse, diabetes, and genetic factors, while in men occurred concomitantly [10].
risk factors include increasing age, other LUTS, In men with benign prostatic hyperplasia, dia-
urinary tract infections, cognitive impairment, betes was associated with greater severity of
neurologic disorders, and prostatectomy [4]. Risk LUTS and lower maximum flow rate [11]. The
factors of overactive bladder in men are age, clinical overlap between benign prostatic hyper-
other LUTS, depression, constipation, neurologic plasia and LUTS and variable definitions of these
conditions, and erectile dysfunction [4]. The conditions might explain conflicting reports of an
presence of LUTS impairs QoL and is associated epidemiological association between benign
with increased risk of depression with a bidirec- prostatic hyperplasia and diabetes [2, 6].
tional relationship between LUTS and both
depression and anxiety [5]. Moreover, in a meta-­ 2.1.2 Bladder Dysfunction in Women
analysis of 15 studies the presence of moderate to In two small studies, abnormal voiding patterns
severe LUTS (without excluding benign prostatic were more common in women with type 2 diabe-
hyperplasia) was associated with major adverse tes than in control women, with a prevalence of
cardiac events in both cross-sectional (odds ratio 13.9% and 22.2% compared to 1.8% and 5.6%
2.38) and longitudinal studies (odds ratio 1.68), and an odds ratio of 4.8 [12, 13]. In three large
with the risk being lower in older patients and population studies, women with diabetes had a
higher in those with diabetes [6]. higher incidence and prevalence of urinary incon-
Epidemiological data on bladder dysfunction tinence compared to those without diabetes (39%
in diabetes are scarce and with wide variations vs. 26%) and an odds ratio between 1.2 and 1.32
reflecting the lack of standardized diagnostic with the greater risk for urge incontinence (up to
modalities and possible selection bias of the stud- 3.5) and for severe forms of incontinence (odds
ied population. Over 50% of women and men ratio 1.54) [14–17]. In the Look AHEAD study in
with diabetes have bladder dysfunction, with overweight/obese women with type 2 diabetes,
bladder instability or hypersensitivity being the weekly incontinence was reported by 27%, with
most frequent finding, ranging from 39% to 61% race/ethnicity (non-Hispanic whites), BMI
of subjects [2]. ≥30 kg/m2, prior hysterectomy, and urinary tract
infection in the prior year being the risk factors
2.1.1 Bladder Dysfunction in Men [18]. In the Diabetes RRISK study in women with
In African American men, a history of diabetes type 2 diabetes at least one urinary symptom was
was associated with an adjusted odds ratio for present in 59% and post-void residual volume
moderate to severe LUTS of 1.95 [7]. In patients ≥100  mL in 12%, with this latter finding being
with type 2 diabetes from Taiwan, 22.5% reported associated with worse glycemic control (odds
overactive bladder [8], with age, male sex, and ratio 1.30 per % increase in HbA1c) [19]. In the
waist circumference being independent risk fac- DCCT/EDIC study at EDIC year 17, urinary
tors for overactive bladder [8]. In the UroEDIC incontinence was present in 30% of women with
study, an ancillary study of DCCT/EDIC study, at type 1 diabetes [20]. Abnormal Valsalva ratio at
EDIC year 17, LUTS were present in 25% of EDIC year 13/14 was independently associated
men with type 1 diabetes [9]. Abnormal Valsalva with increased risk of developing urinary inconti-
ratio at DCCT closeout independently predicted nence at EDIC year 17 (odds ratio 1.68), while at
the presence of ED and LUTS at EDIC year 17 EDIC year 17 urinary incontinence was associ-
(odds ratio 1.50) [9]. At EDIC year 17, LUTS ated with age and BMI ≥30 kg/m2 [10, 20]. In the
were associated with increasing age, micro- same study, HbA1c predicted the development of
(odds ratio 1.86) and macroalbuminuria (odds urinary incontinence from year 10 to year 17 with
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 493

an adjusted odds ratio of 1.41 per % increase in 2.2 Physiology of Micturition


mean HbA1c in EDIC years 1–10 [21].
The lower urinary tract is composed of the blad-
2.1.3 Urinary Tract Infections der and the urethra, which are responsible for
In a large population-based German study, the urine storage and voiding. Micturition is regulated
incidence of urinary tract infections in patients by a complex neural circuitry involving the
with type 2 diabetes was 111.8/55.8 per 1000 peripheral and central nervous system. The blad-
patient-years for women/men, with age der is mainly innervated by the parasympathetic
>79 years, female gender, history of urinary tract pelvic nerve, while the urethra by the sympathetic
infections and cystoscopy, number of comorbidi- hypogastric nerve and somatic pudendal nerve.
ties, HbA1c >9% and GFR <60  mL/min being Bladder storage depends on the sympathetic acti-
associated with increased risk [22]. A meta-anal- vation of β3-adrenoceptors and simultaneous
ysis of 22 studies documented an odds ratio for deactivation of parasympathetic M2-M3 musca-
asymptomatic bacteriuria of 3.0 and 3.2 in type 1 rinic receptors, resulting in bladder relaxation.
and type 2 diabetes, respectively, and of 2.6 and Closure of the bladder neck is due to the sympa-
3.7  in women and men, respectively [23]. thetic activation of α1A/D adrenoceptors. Storage
Moreover, in patients with diabetes a greater risk function is maintained by the spinal cord inhibi-
of developing a symptomatic urinary infection, tory reflex, which is facilitated by the pontine
of having unusual infecting organisms, and of continence center and also by the hypothalamus,
progressing to serious or uncommon conse- cerebellum, basal ganglia, and frontal cortex.
quences was described [2, 24]. Postmenopausal Bladder voiding depends on a spino-­bulbo-­spinal
state and diabetes treatment complexity were excitatory reflex that is triggered by afferents
associated with a higher risk of urinary tract fibers from tensor receptors in the bladder and
infections in type 2 diabetes, whereas in type 1 involves the periaqueductal gray in the midbrain
diabetes sexual activity and not duration of dia- and the pontine micturition center. Fibers project-
betes or HbA1c increased the risk [2]. In the ing from pontine micturition center activate the
DCCT/EDIC study, urinary tract infection, pres- sacral bladder preganglionic nucleus responsible
ent in 17% of women with type 1 diabetes, was for bladder contraction (through M2-M3 musca-
associated with microalbuminuria (odds ratio of rinic receptors of acetylcholine) and inhibit the
2.08) and CAN (odds ratio of 1.57) [10, 20]. sacral urethral motor nucleus (Onuf’s nucleus),
In summary, bladder dysfunction findings are causing striated sphincter relaxation. The frontal
present in >50% of people with diabetes, with cortex and hypothalamus initiate the bladder
bladder instability being the most common. Men voiding reflex and are therefore involved in both
with diabetes have a twofold risk of LUTS and a storage and voiding [25]. Due to this complexity,
prevalence of storage LUTS of 22.5% in type 2 bladder function is sensitive to numerous injuries
diabetes and of any LUTS of 25% in type 1 diabe- or diseases affecting the nervous system.
tes. Risk factors are age in type 2 diabetes and
age, microalbuminuria, and CAN in type 1 diabe-
tes. The most common urinary symptom in women 2.3 Pathophysiology of Diabetic
is incontinence present in 27% to 39% in type 2 Bladder Dysfunction
and in 30% in type 1 diabetes, with an odds ratio
from 1.2 to 3.5. Risk factors for incontinence in Expectedly, diabetic damage of both somatic
type 2 diabetes are obesity, ethnicity, hysterec- and autonomic fibers was first considered as the
tomy, and urinary infection, while in type 1 diabe- main pathogenetic mechanism of diabetic blad-
tes age, BMI, glycemic control, and CAN. Urinary der dysfunction underlying its classical form of
infections are more common in people with diabe- diabetic cystopathy, through the involvement of
tes, with diabetes being a promoter of their pro- (1) sensory afferent fibers responsible for
gression. In most men with type 1 diabetes LUTS decreased bladder sensation and increased
overlap with sexual dysfunction. bladder capacity and (2) parasympathetic effer-
494 V. Spallone et al.

ent fibers leading to impaired detrusor contrac- bladder pain, while reduced NGF is a conse-
tility and reduced bladder emptying. However, quence of a deficiency in bladder production or
urodynamic and animal studies have suggested nerve transport to the bladder [33]. Moreover,
a spectrum of different facets to bladder dys- NGF gene therapy by herpes-virus-mediated
function and a multifocal influence of diabetes. NGF gene delivery into the bladder prevented
Moreover, they have founded a temporal theory excessive increases in bladder capacity and
of diabetic bladder with a progression from an reduced post-void residual volume in diabetic
early compensated storage phase characterized rats [32]. In addition, the bladders of diabetic
by bladder hypertrophy and overactivity and a mice showed an increase in the proNGF/NGF
late decompensated voiding phase with detru- ratio and in the coexpression of proNGF and p75
sor underactivity [26]. However, a clear demon- neurotrophin receptor (p75NTR) in the mucosa
stration of this phase progression in humans is in association with high levels of tumor necrosis
lacking, not all the studies fill in this framework factor-α and bladder changes, which were
[27] and this view might turn out to be overly reverted by the inhibition of proNGF and antago-
simplistic. nism of p75NTR [34]. These findings suggest an
involvement of NGF and the proNGF/p75NTR
2.3.1 Diabetic Neuropathy axis in bladder dysfunction pathogenesis.
Diabetic autonomic neuropathy and polyneurop-
athy have been associated with LUTS and uri- 2.3.3 Detrusor Smooth Muscle
nary incontinence in type 1 diabetes [9, 20], as Dysfunction
well as diabetic polyneuropathy being associated In animal models of diabetes, early adaptive
with low flow rate on urodynamic evaluation modifications of detrusor muscle have been
and, somewhat controversially, with high post- described: changes in biological pathways and
void residual in type 2 diabetes [12, 19, 27]. up- and downregulation of proteins involved in
Abnormal electrophysiologic findings were muscle function including the proapoptopic path-
shown in patients with diabetes and LUTS, i.e., way, altered membrane lipid composition and
peripheral neuropathy in urethral striated sphinc- neurotransmitter release, enhanced muscarinic
ter and bulbo-cavernosus muscles, increased sensitivity, abnormal purinergic transmission,
latency of bulbo-cavernosus reflexes, and and hyperglycemia-driven pathways suggesting
delayed somatosensor evoked responses of tibial elevated oxidative stress and tissue remodeling
and pudendal nerves [28, 29]. In addition to the [33, 35]. Chronic administration in mice of meth-
finding of the reduced density of acetylcholines- ylglyoxal, a product of glycolysis involved in the
terase positive staining nerves in the bladder advanced glycation end products (AGEs) forma-
wall in patients with severe type 1 diabetes [30], tion, induced bladder inflammation and increased
an impairment in Aδ and C fiber bladder afferent micturition frequency and number of non-­voiding
pathways was found in women with type 2 dia- contractions at the urodynamic evaluation, sug-
betes using urodynamics with intravesical cur- gestive of an overactive bladder [36].
rent perception threshold testing, which was
associated with poor emptying function and 2.3.4 Urothelial Dysfunction
detrusor hypoactivity [31]. Urothelial cells have sensor molecules and,
when activated by mechanical or chemical
2.3.2 Neurotrophins stimuli via these receptors and ion channels,
Studies in diabetic animals have shown an they can in turn release mediators, such as nitric
increase in bladder nerve growth factor (NGF) oxide (NO), ATP, acetylcholine, prostaglan-
levels in the early compensatory phases and a fall dins, and substance P and thus interact with
in the decompensated phases of bladder dysfunc- nerves endings, interstitial cells, smooth mus-
tion [32] leading to the hypothesis that increased cle, and blood vessels with an amplifying effect
NGF is involved in detrusor overactivity and of stimuli [32, 35]. Chronic hyperglycemia can
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 495

alter receptor expression and cause a breach of potentially leading to increased outlet resistance
the barrier and may influence afferent nerves, and voiding dysfunction [39]. The urethra is sub-
thus contributing to bladder instability, hyper- ject to time-dependent changes as with bladder:
activity, and altered bladder sensation [35]. A in the early stage increased urethral pressure dur-
diabetes-induced urothelial senescence has ing micturition, subsequent detrusor-sphincter
been also hypothesized, capable of both dis- dyssynergia, and finally reduced thickness of the
rupting the barrier function and altering signal- urothelium and atrophy of striated muscle bundle
ing and sensation of bladder fullness, resulting impairing voiding efficiency [40] (Fig. 1).
in overactive symptoms [37]. Finally, adher- In summary, somatic and autonomic neurop-
ence of Escherichia Coli with type 1 fimbriae to athy play a major role in bladder dysfunction,
the uroepithelial cells from women with diabe- mainly involving voiding function, but other
tes was twofold compared to controls and was diabetes-­related mechanisms directly affect
related to HbA1c, suggesting an effect of gly- bladder and urethra. The traditional model of
cosylation on bacteria receptors [38]. diabetic cystopathy, including neuropathy as
the main culprit, has been replaced by the con-
2.3.5 Urethra Dysfunction cept of a multilevel spectrum of mechanisms,
In diabetic rats, a condition of urethropathy has and a two-­phase model of bladder dysfunction
been described including external urethral (Fig. 1). Pathophysiology of diabetic bladder
sphincter dysfunction, decreased NO-induced remains, however, incompletely explored
relaxation and instead sensitization to α1-­ together with differences between type 1 and
adrenergic agonists of urethral smooth muscle, type 2 diabetes.

Early Late
Storage Phase Voiding Phase

Factors Polyuria Factors Hyperglycaemia (oxidative stress)


Neuropathy
↓ Neuropeptides
Changes ↑ Bladder mass Detrusor changes
↑ Urethra tickness Bladder-sphincter incoordination
Instrumental Detrusor Overactivity Changes ↓ Urothelium thickness
findings ↓ Striated muscle

Symptoms Urgency, incontinence, Instrumental Detrusor Underactivity


frequency, nocturia findings ↑ post-void residual volume
Possible concomitant BPH (in
men)
Symptoms ↓ sensation, difficulty in voiding
(weak stream, dribbling),
sensation of incomplete voiding

Fig. 1  A temporal progression model of diabetic bladder stage, hyperglycemia-driven pathogenetic mechanisms,
dysfunction. A two-phase temporal progression model of like oxidative stress, induce structural changes in smooth
bladder dysfunction has been proposed. In an early phase, muscle cells of detrusor and the urethra, including apopto-
polyuria consequent to hyperglycemia would promote an sis [41], neuropathy and neurodegeneration in the bladder
adaptive remodeling of the bladder and urethra, including [42], and possibly changes in urethro-vesical crosstalk
hypertrophy, while other functional changes regard neuro- [40]. This stage of decompensated bladder function is
nal transmitter release and excitability and NO production characterized by an underactive bladder and urethra,
and control with increased protein nitration [33]. Bladder decreased micturition pressure, and increased post-void
and urethra overactivity and storage LUTS are the corre- residual with the emergence of voiding symptoms [35]
spondent instrumental and symptomatic findings. In a late
496 V. Spallone et al.

2.4 Clinical Manifestations factor for symptomatic infections, complications,


of Diabetic Bladder and antibiotic resistance [44]. Due to their mech-
Dysfunction anism of action, SGLT2i have been associated
with urinary infections with a relative risk of
LUTS in patients with diabetes are often indolent 1.29  in a meta-analysis of seven clinical trials
and unrecognized despite the impact on QoL and [45], although a subsequent meta-analysis did not
the prognostic meaning. In the early stage, confirm this finding and also disproved a dose–
urgency, increased daytime frequency, and noctu- response relation between most SGLT2 inhibi-
ria constitute the reason for an early urologic tors and urinary infections with the exception of
referral, as well as urinary incontinence, while dapagliflozin [46]. However, conclusions cannot
voiding symptoms are generally later complaints be made regarding serious urinary infections as
(Fig. 1). pyelonephritis, which a safety announcement of
the Food and Drug Administration (FDA)
2.4.1 Manifestations in Men reported as associated with SGLT2i [47].
and Women
The distribution of LUTS differs between women
and men, with a predominance of an overactive 2.5 Diagnosis of Diabetic Bladder
bladder in men and of incontinence in women. In Dysfunction
men, bladder storage symptoms and voiding
symptoms might be related to both benign pros- Diagnostic pathway includes a preliminary
tatic hyperplasia and diabetic bladder dysfunc- assessment and a specialist neuro-urological
tion, often coexisting in men over 50, making assessment (Fig. 2).
their differential diagnosis challenging in the pri-
mary care setting. 2.5.1 History, Questionnaires,
In women, symptoms are increased daytime and Diaries
frequency, nocturia, but also weak urinary Patients history is of paramount importance in the
streams and incomplete voiding with increased initial diagnostic evaluation of LUTS and should
post-void residual [12, 13, 19]. This latter finding include: (1) BMI, diabetes history; (2) lifestyle
in the Diabetes RRISK study was associated with and habits (smoking, alcohol, addictive drugs),
both obstructive voiding symptoms (2.47 odds the number of absorbent pads used per day, QoL,
ratio) and urge incontinence (2.18 odds) [19]. disabilities; (3) comorbidities, m
­ edication history
However, the most common symptom in women (opioids, anticholinergic agents, α-adrenoceptor
is urinary incontinence [14]. While in type 2 dia- agonists); (4) surgical history (abdominopelvic
betes urge incontinence is the most frequent form and urologic surgery), and in women parity,
[14, 16], in type 1 diabetes mixed or stress incon- obstetric and gynecological surgery; (5) a detailed
tinence seems to be prevalent [21]. investigation of LUTS with regard to onset, initia-
tion (hesitancy, straining, Credé maneuver), and
2.4.2 Urinary Tract Infections interruption (paradoxical, passive) of micturition,
Diabetes causes a threefold risk of asymptomatic voiding frequency, nocturia, urgency, stress,
bacteriuria independent of glycemic control, with urgency or mixed urinary incontinence; (6) mac-
pooled prevalence of 14.2% in women and 2.3% rohematuria and signs of urinary infections; (7)
in men [23]. Preliminary treatment of asymptom- genital, sexual, and bowel dysfunction symptoms.
atic bacteriuria does not reduce the risk of symp- Standardized questionnaires validated in different
tomatic infections and complications and it is not languages represent a useful tool to quantify
recommended by scientific societies together symptoms severity, including: (1) the International
with its screening [43, 44]. Nonetheless, diabe- Prostatic Symptoms Score (IPSS), validated in
tes, mainly in presence of poor glycemic control men with benign prostate hypertrophy, allows for
and high post-void residual, is a host-related risk easy assessment of LUTS (excluding inconti-
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 497

Preliminary diagnostic assessment


History, questionnaires and bladder diary
Physical examination
Urinalysis +/- urine culture and eGFR
Uroflowmetry and post-void residual measurement (ultrasound)

Symptomatic urinary tract Overactive bladder ± Urgency Stress urinary incontinence Voiding-phase symptoms
infection incontinence Lifestyle intervention Lifestyle intervention
Treat infection Lifestyle intervention Pelvic floor muscle training α1-blockers (in men)
Repeat preliminary assessment Antimuscarinics or β-3 agonists Duloxetine Intermittent catheterization
(Do not treat asymptomatic
bacteriuria)

Yes Are symptoms No


refractory to 1st
line therapy?

2nd level diagnostic and specialist Confirm 1st line therapy


neurourological assessment Follow-up of symptoms and QoL using
Urodynamics questionnaires
+/- Urethrocystoscopy Repeat preliminary assessment if
+/- Upper and Lower urinary tract ultrasound symptoms change
+/- Neuro-urophysiological tests

Refractory detrusor overactivity +/- Stress urinary BPH-related symptoms


Detrusor underactivity
incontinence incontinence Stepwise therapeutic approach:
Stepwise therapeutic
Stepwise therapeutic approach: Surgery tailored to Associate 5-ARIs when
st
approach:
Titrate dose of 1 line therapy patient-specific factors indicated
Intermittent catheterization
Switch to or combine with 2nd line therapy BPH surgery
Tibial nerve stimulation
Tibial nerve stimulation Sacral neuromodulation
Onabotulinum A toxin Injection
Sacral neuromodulation
Augmentation cystoplasty
Urinary diversion
Periodical specialist symptom and QoL reassessment is always advisable in patients with symptoms refractory to 1st line therapy. Therapeutic approach in patients with mixed presentations
(i.e., urgency and detrusor underactivity, stress and detrusor overactivity incontinence) may vary and require expert neuro-urological assessment.
Consider screening of diabetic polyneuropathy and autonomic neuropathy.

Fig. 2  An algorithm for a diagnostic and therapeutic to plan surgery. Upper urinary tract ultrasound and ure-
approach to diabetic bladder dysfunction. A preliminary throcystoscopy are reserved for differential diagnosis
assessment in a patient with diabetes and LUTS is based [50–52]. The first-line treatment includes lifestyle inter-
on history, questionnaires, bladder diary, urinalysis, uro- ventions for all LUTS and antimuscarinics or β-3 agonists
flowmetry, and post-void residual measurement (by supra- for overactive bladder, pelvic floor muscle training and
pubic ultrasound). This relatively inexpensive approach duloxetine for stress incontinence, and intermittent cathe-
often supplies all of the information needed. terization for voiding symptoms. In men with bladder out-
Questionnaires and a bladder diary at scheduled intervals let obstruction α1-blockers can be used. In the event of
(i.e., 6 to 12-monthly) allow for the monitoring of treat- inefficacy, when a definite diagnosis is obtained, an
ment outcomes [48, 49]. Uroflowmetry and post-void upgrading of treatment for detrusor overactivity includes
residual measurement can be repeated as well to monitor combination of first-line drugs, tibial nerve stimulation,
the urodynamic treatment effects or in the event of clinical onabotulinumtoxinA, sacral neuromodulation and, as a
changes. Urinalysis with urine culture should verify the last resort, surgery. A similar stepwise approach is applied
efficacy of antibiotic treatment or the suspicion of new for detrusor underactivity with tibial nerve stimulation
infection. Invasive tests like urodynamics are advisable and sacral neuromodulation [53–56]. eGFR estimated
only when the preliminary approach is inconclusive or the glomerular filtration rate, BPH benign prostatic hyperpla-
initial symptom-based treatment is ineffective or in order sia, 5-ARIs 5-α reductase inhibitors

nence) and LUTS-­related QoL; (2) the Overactive of preliminary assessment and follow-up of
Bladder Questionnaire Short Form (OAB-q SF) LUTS.  It is a semi-objective tool that quantifies
and the International Consultation on Incontinence LUTS and urodynamic variables such as every
Questionnaire-Urinary Incontinence Short Form voided volume, nocturnal, and daytime voided
(ICI-Q UI SF) are focused on urgency symptoms, volume, for a recommended duration of at least
incontinence and related QoL.  Gender specific 3  days [48, 49]. Patient’s motivation can favor
extended versions of the ICI-Q are also available accurate data collection.
(ICIQ-MLUTS and ICIQ-FLUTS) [57, 58]. A Given the insidiousness and possible patient
bladder/voiding diary is an essential component unawareness of abnormal micturition patterns,
498 V. Spallone et al.

questions on urinary symptoms should be mated through a suprapubic ultrasound scan.


included in the yearly assessment of people with Results of uroflowmetry and post-void residual
diabetes together with urine analysis. are needed to plan medical or surgical treatment
Questionnaires for autonomic symptoms such as of LUTS [51]. While suprapubic ultrasound can
Compass 31 [59] contain questions on inconti- be integrated in the uroflowmetry to estimate
nence and difficult and incomplete voiding and post-void residual, upper urinary tract imaging is
might be useful for a preliminary identification of not necessary in the preliminary assessment
these symptoms, but a comparison with specific unless indicated by clinical features, taking into
questionnaires is lacking. account that patients with neurogenic LUTS may
develop urinary tract stones [50].
2.5.2 Physical Examination
Physical examination constitutes an essential 2.5.5 Invasive Urodynamics
bedside evaluation of patients with LUTS, includ- and Urethrocystoscopy
ing: (1) in males, abdominal, inguinal, genital Urodynamic studies are minimally invasive tests
examination to rule out acute urinary retention or in the outpatient setting that complete the diag-
infections, mass or alteration of the external ure- nostic pathway of LUTS, guide treatment
thral orifice, digital rectal examination as part of choices, and assist in patient’ counseling. A
a standard neuro-urological assessment; (2) in combination of multichannel filling cystometry
females, alongside abdominal and pelvic exami- and pressure/flow study is generally used in
nation, vaginal examination to assess estrogen patients with diabetes. Urodynamics assesses
status, pelvic organs prolapse, and urethral hyper- detrusor and bladder outlet function and can lead
mobility  with or without  stress urinary inconti- to the diagnosis of urodynamic incontinence or
nence during the cough test. When appropriate, a severe detrusor overactivity in the absence of
more focused neuro-urological exam should incontinence, can suggest treatment in patients
investigate sensation in the S2-S5 dermatomes resistant to first-line therapy, and can help to dif-
bilaterally, the bulbocavernosus and perianal ferentiate the cause of bladder outlet obstruction
reflex, anal sphincter tone and voluntary contrac- symptoms in males (due to either benign pros-
tion, and pelvic floor muscle maximal and tatic hyperplasia, detrusor underactivity or both)
repeated contractions with the pubococcygeal [51]. Invasive urethrocystoscopy can be suitable
test [50]. for the diagnosis of bladder outlet obstruction
(i.e., urethral stenosis) and in the event of urethra
2.5.3 Laboratory Investigations or bladder pathologies are suspected (i.e., gross
Urinalysis is always recommended to detect hematuria).
signs of infection, together with urine culture and
antibiotic sensitivity tests in the presence of 2.5.6 Uro-Neurophysiological Tests
symptomatic urinary infections. Kidney function Electrophysiological testing of the motor or sen-
is usually included in the monitoring of diabetic sory function of the pelvic floor muscles, urethral
nephropathy. and anal sphincter, pudendal nerves, bladder, and
urethra is available and able to identify peripheral
2.5.4 Instrumental Diagnostic neuropathy, but they are considered elective pro-
Uroflowmetry with measurement of post-void cedures [52].
residual is a non-invasive test that is necessary in Given the association between lower urinary
the preliminary evaluation and requested prior to tract dysfunction with CAN and polyneuropa-
any further invasive test in patients with LUTS thy, screening for these complications, through
who are able to void. Uroflowmetry measures the assessment of sensorimotor and autonomic
voided volume and Qmax. Post-void residual is symptoms and signs, is advisable in patients
measured by catheterization or preferably esti- with diabetes and LUTS and vice versa screen-
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 499

ing for LUTS in those with diabetic neuropathy shown efficacy on urinary incontinence (but not
[29, 60]. nocturia or frequency) also in overweight/obese
A stepwise algorithm for diagnostic manage- men with type 2 diabetes in the Look AHEAD
ment of patients with diabetes and LUTS is illus- trial with almost double the odds of urinary
trated in Fig. 2. incontinence resolving in the intensive lifestyle
intervention group compared to only diabetes
support and education [62]. Pelvic muscle train-
2.6 Treatment of Diabetic Bladder ing can improve symptoms of stress and other
Dysfunction forms of urinary incontinence and should be con-
sidered in first-line conservative management
Treatment is multimodal and focuses on the pres- program for patients with urinary incontinence
ervation of renal function, relief of LUTS, pro- [52, 54].
motion of continence, prevention of infectious
complications, and amelioration of QoL [53]. 2.6.2 Treatment of Symptoms
Management strategies include behavioral inter- Related to Benign Prostatic
vention, pharmacological treatment, and surgery. Hyperplasia
Patient reported outcomes and diaries are useful Men with diabetes presenting with symptoms of
in the follow-up to monitor changes and treat- bladder outlet obstruction might benefit from
ment outcomes. α1-adrenoceptor antagonists as first-line therapy,
with a preference for selective α1-blockers (tam-
2.6.1 Lifestyle Interventions sulosin, silodosin) and similar efficacy as in men
General behavioral modifications are advised in without diabetes [11]. Drug-induced vasoactive
any patient presenting with LUTS.  A reduction effects could exacerbate orthostatic hypotension
of fluid intake in certain day periods might help in those with CAN. Treatment with 5-α reductase
to control nocturia. Scheduled time voiding inhibitors (5-ARIs) reduces acute urinary reten-
might be employed in the presence of decreased tion episodes and the need for surgery and is con-
bladder sensitivity and high voided volumes. sidered in association with α1-blockers in patients
Reduction of alcohol and caffeine intake might with prostate volumes >40  mL and prostate-­
prevent diuretic effects and their irritative conse- specific antigen levels >1.4–1.6 ng/mL. Specialist
quences. A review of medications and their time evaluation and prostate cancer screening, assess-
of administration (especially diuretics) is advis- ment of sexual function, and proper counseling
able to reduce bothersome effects on LUTS.  In on the possible impact on sexual function are
addition to glycemic control, whose association advisable in patients with diabetes prior to pre-
with incontinence in type 1 diabetes [21] and scription [63]. When indicated, surgery can be
voiding function in type 2 diabetes [19] is well performed, taking into account in preoperative
documented, lifestyle interventions play a spe- counseling that diabetes is associated with some
cific therapeutic role in patients with urinary persistence of LUTS and voiding dysfunction at
incontinence. Results from the DPPOS study 3 months after surgery [64].
showed a lower prevalence of urinary inconti-
nence in overweight women with impaired glu- 2.6.3 Treatment of Urgency, Detrusor
cose tolerance enrolled in intensive lifestyle Overactivity, and Urgency
intervention group compared to oral metformin Incontinence
and placebo groups over the course of a 6-year Muscarinic receptor antagonists (antimuscarin-
follow-up period (46.7% vs. 53.1% vs. 49.9% ics) are the cornerstone of treatment of storage
urinary incontinence/week) with a mediating symptoms in the general population and can be
effect of reduction in BMI and waist circumfer- also offered to patients with diabetes. All avail-
ence [61]. Intensive lifestyle intervention has able formulations have similar clinical efficacy
500 V. Spallone et al.

[65]. M3 selective inhibitor darifenacin has term durability of treatment [69] and on the
shown similar efficacy in patients with overac- efficacy in patients with diabetes are needed.
tive bladder and diabetes vs. those without Implantation of a permanent sacral neuromodu-
comorbidities, and a similar class effect for all lator (SNM) seems to modulate spinal cord
antimuscarinics might be suggested [65]. reflexes and brain networks by peripheral affer-
Patients’ counseling on side effects like xerosto- ents. It was similarly effective in patients with
mia, constipation, dizziness, blurred vision, diabetes as in those without in a 29-month fol-
nasopharyngitis, and occasionally tachycardia low-up study, with ­success rates of 69.2% for
is advisable, as well as on the possible worsen- urgency urinary incontinence and 85.7% for
ing effect on post-void residual, which requires urgency-frequency [70]. The finding of a higher
regular voiding symptoms monitoring and number of explants due to infection in patients
1-month re-­evaluation and represents a contra- with diabetes in this study (16.7% vs. 4.3%)
indication if ≥150 mL. Combination therapy of [70] was not however confirmed in another
antimuscarinics with α1-blockers can be effec- study (3.1% vs. 6.8%) [71]. While studies com-
tive in men with moderate-severe LUTS not paring TNS and SNM are lacking, lower risk
responding to a single first-line drug, taking into profile, minimally invasive nature, and lower
account the safety profile of both. Mirabegron is (initial) costs might favor TNS, although SNM
the only licensed oral β-3 adrenoceptor agonist was more intensively investigated. Patient pref-
with efficacy in treating voiding frequency, erence and compliance are other aspects to con-
urgency, and urgency incontinence in the gen- sider when choosing the neuromodulation
eral population. A pooled analysis of trials procedure.
showed a better tolerability profile compared to OnabotulinumtoxinA (onabotA) injection in
antimuscarinics in elderly patients [66], with the detrusor muscle represents a well-established
less xerostomia and constipation. However, therapeutic alternative for refractory detrusor
hypertension, headache, and nasopharyngitis overactivity and urgency incontinence both in
are drug specific adverse effects, while severe the idiopathic and neurogenic forms. The proce-
uncontrolled hypertension is a contraindication. dure is minimally invasive (by cystoscopy under
Evidence on combination therapy with mirabe- local anesthesia) and can be performed in the
gron is lacking. outpatients setting. In a retrospective study, ona-
Electrical neuromodulation is a therapeutic botA obtained in 48 patients with diabetes and
option in patients with an overactive bladder. refractory detrusor overactivity a similar success
Tibial nerve stimulation (TNS), via a percuta- rate as in patients without diabetes (56% vs.
neous or transcutaneous approach, delivers 61%), but those with diabetes had a higher
electrical stimuli to the sacral micturition cen- 6 month rate of large post-void residual and of
ter via the S2-S4 sacral nerve plexus through a general weakness [72]. Thus, patients should
fine gauge needle or a patch positioned above receive preliminary information on the drug
the medial malleolus. Treatment is performed safety profile and adequate training in clean
in an outpatient setting in cycles of 10–12 once- intermittent catheterization if motivated to start
weekly stimulations of 30 min. TNS was effec- treatment.
tive and comparable to the antimuscarinic drug Augmentation cystoplasty is another option to
tolterodine in females with urgency urinary treat refractory detrusor overactivity. No data are
incontinence [67, 68] and also effective in those available on comparison of this alternative with
not benefiting from antimuscarinics. TNS is a SNM or onabotA injection in the general or dia-
minimally invasive and safe option available as betic population. Urinary diversion is the last sur-
a second therapeutic line or in combination gical choice to treat symptoms that are refractory
with oral treatment. However, data on the long- to any other therapeutic approach.
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 501

2.6.4 Treatment of Detrusor stomia, constipation, dizziness, insomnia, som-


Underactivity nolence, and fatigue and can lead to premature
Despite preclinical studies with cholinergic drugs discontinuation [75]. Surgical treatment includes,
and intravesical cannabinoids, no pharmacologi- depending on the underlying cause of inconti-
cal treatment has been validated for detrusor nence, adjustable continence therapy balloons,
underactivity. Clean intermittent self- or aided-­ urethral slings, artificial urinary sphincter, and
catheterization still represents the preferred other open or laparoscopic assisted incontinence
option for patients with underactive detrusor and surgeries with progressively increasing invasive-
high post-void residual or no detrusor contractil- ness. The surgical procedure should be tailored to
ity. Treatment reduces bacteriuria and infections the patient and performed by expert functional
compared to indwelling catheterization [73]. urologist in referral centers. As for other pros-
Frequency of daily catheterizations should be tai- thetic implants, diabetes and obesity were associ-
lored to symptoms and degree of underactivity ated with mesh erosion for trans-vaginal and
and to maintain voiding volumes less than 400– trans-obturator sling surgery [76]. Stress urinary
500 mL. Patient education on technique is essen- incontinence rarely occurs in males unless as a
tial and a third-party catheterization for patients consequence of prostatic surgery. Surgical
with poor manual dexterity or cognitive dysfunc- options are similar in the general and diabetic
tion is a preferable option to indwelling catheter- populations and include balloons, slings, and
ization. In a systematic review, TNS showed artificial sphincter implantation.
limited efficacy in patients with idiopathic non-­ Figure 2 presents a therapeutic algorithm for
obstructive urinary retention with an objective bladder dysfunction in diabetes.
success rate (reduction in number of catheteriza-
tions or catheterized volume) up to 41% and a
subjective success rate (patient’s request for con- 2.7 Barriers to Effective
tinued chronic treatment) up to 59% [55]. No Management of Diabetic
studies are available in patients with diabetes. Bladder Dysfunction
SNM represents a therapeutic option in patients
with diabetes and detrusor underactivity with a Despite significant advancement in the last two
success rate of 66.7% [70] but with a possible decades, the natural history and pathophysiology
increased risk of infection. SNM might enable of diabetic bladder dysfunction are still poorly
simultaneous pancreas and kidney transplanta- known. It remains underrepresented in research,
tion in candidate patients with detrusor acontrac- epidemiological studies do not adopt standard-
tility requiring clean intermittent catheterization ized diagnostic criteria and are most cross-­
and at high risk of recurrent urinary infections by sectional, therapeutic trials are very few, thus
restoring voiding and preventing urinary infec- limiting the possibility to build treatment evi-
tions [74]. dence, with data on tolerability and treatment
adherence very limited. Cost-effectiveness stud-
2.6.5 Treatment of Stress Urinary ies on diagnostic and treatment pathways are
Incontinence lacking and awareness of bladder dysfunction is
The selective serotonin-norepinephrine reuptake not widespread in clinical practice. Diagnosis
inhibitor (SNRI) duloxetine increases the stimu- often occurs at an advanced stage, and manage-
lation of the spinal cord receptors of the pudendal ment is almost exclusively in the hands of urolo-
motor neurons and improves the resting tone and gists [56]. Nonetheless, the evidence of a
contraction strength of the striated urinary beneficial role of the lifestyle intervention, of
sphincter. Duloxetine was found to improve glycemic control and even of the choice of anti-
female stress urinary incontinence and related hyperglycemic drugs [77] as well as the prognos-
QoL. Side effects are nausea and vomiting, xero- tic cardiovascular meaning of the LUTS in males
502 V. Spallone et al.

[6], points to the need for a holistic and multidis- those without [80]. Moreover, although related to
ciplinary approach to the prevention and manage- age and diabetes duration, ED starts early in dia-
ment of diabetic bladder dysfunction (including betes, might be present in 32% of patients with
the general practitioner, urologist, endocrinolo- recently diagnosed type 2 diabetes, as docu-
gist, neurologist, gynecologist, and cardiologist). mented by SUBITO-DE study [83], and may be
associated with undiagnosed diabetes, as in the
NHANES study, with an odds ratio of 2.2 that
3 Male Sexual Dysfunction reached a value of 8.7  in the 40–59 age group
in Diabetes [84]. The MMAS provided an incidence rate of
ED of 26 cases/1000 man-years [85]. In Italian
Male sexual dysfunction includes ED, abnormal- men with diabetes the incidence of ED was 68
ities of orgasmic, ejaculatory function, sexual cases/1000 person-years, more than twice that
desire, enjoyment, and failure of genital response. seen in the MMAS [86].
Although diabetes may well affect different In summary, both the prevalence and inci-
aspects of sexual health, most attention has been dence of ED are more than tripled and doubled,
focused on ED. respectively, in males with diabetes compared to
healthy people. ED develops 10–15 years earlier
in men with diabetes. ED starts early in diabetes
3.1 Epidemiology of Diabetic and can be present at diabetes diagnosis or even
Erectile Dysfunction precede it.

3.1.1 Prevalence of Diabetic Erectile 3.1.2 Clinical Correlates,


Dysfunction Comorbidities, and Risk Factors
According to the Fourth International of Diabetic Erectile Dysfunction
Consultation on Sexual Medicine [78] ED is Population-based studies have shown that in
defined as the consistent or recurrent inability to addition to age, clinical correlates or comorbidi-
attain and/or maintain penile erection sufficient ties of ED were pelvic surgery, diabetes (an age-­
for sexual satisfaction. In the population-based adjusted odds ratio of 3.95), LUTS, hypertension,
Massachusetts Male Aging Study (MMAS), the heart disease, heavy smoking, and depression,
prevalence of moderate and severe ED was 35% with instead a protective role of education, physi-
[79]. Many studies documented an association cal activity, and alcohol drinking [87, 88].
between ED and diabetes. A meta-analysis of 145 Clinical correlates of ED in diabetes were found
studies showed a prevalence of ED of 59.1% in to be advancing age, diabetes duration, poor gly-
diabetes, of 37.5% in type 1 diabetes and of cemic control, hypertension, hyperlipidemia,
66.3% in type 2 diabetes [80]. This meta-analysis sedentary lifestyle, smoking, and diabetic com-
highlighted the heterogeneity of the included plications [89]. However, in the cited meta-­
studies with regard to the prevalence rate (from analysis by Kouidrat et al. [80] only hypertension
35% to 80%), the countries, the diagnostic was a moderator of ED presence in men with dia-
approach, and the information on diabetic com- betes. When considering longitudinal studies, in
plications. In the eight studies comparing men the MMAS, lower levels of education, diabetes,
with diabetes and healthy controls, the preva- heart disease, and hypertension were indepen-
lence was 51.6% and 25.5%, respectively, with dent predictors of ED [85], while in people with
an odds ratio for ED of 3.62 [80]. In a large diabetes, age, diabetes duration, renal disease,
Italian cross-sectional study, the mean prevalence and hypertension played a predictive role for ED
of ED in men with diabetes was 35.8% [81], [86]. In the UroEDIC II study, ED was present in
compared to 12.8% in the general population 45% of men with type 1 diabetes and in more
[82]. Available data suggest that ED develops in than half the cases it was associated with other
people with diabetes 10–15 years earlier than in urological complications, with age, poorer glyce-
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 503

mic control, and other diabetic complications QoL [95]. In the UroEDIC study, at EDIC year
including CAN [10]. Moreover, during a 16-year 17, the presence of ED was associated with worse
follow-up the rate of incident ED was 43.4 cases QoL (an odds ratio of 3.01) and higher level of
per 1000 person-years [90]. Risk of ED increased psychiatric symptoms independently of depres-
by 21% with each 10 mmHg of systolic BP eleva- sion or microangiopathic complications [96].
tion but only in those not under antihypertensive Moreover, men with history of effective treat-
medications [90]. Similarly, in type 2 diabetes in ment of ED had almost identical QoL as those
the Look AHEAD trial, at a cross-sectional level, who had never experienced ED [96]. Possible
every 10 mmHg increase in systolic/diastolic BP mechanisms linking depression and ED in diabe-
was associated with a 30%/10% increased preva- tes are both biological and psychological and
lence of ED [91]. might also include hypogonadism, autonomic
In summary, in men with diabetes, clinical dysfunction, and the presence of comorbidities.
correlates of ED are age, diabetes duration, gly- In summary, in the diabetic scenario where
cemic control, hypertension, hyperlipidemia, depression and diabetes already have a recipro-
sedentary lifestyle, smoking, diabetic complica- cal relationship, the presence of ED increases the
tions, and LUTS, with age, duration, glycemic risk of depression and vice versa, the association
control, hypertension, and renal disease being might be present at diagnosis of type 2 diabetes.
longitudinal predictors of ED, and with an over- ED has an adverse impact on QoL, independent
all prominent role of hypertension. of depression, with a tripled risk of having low
QoL in type 1 diabetes. The relationship, also in
Depression and QoL this case, is bidirectional given that ED predicts
In the general population, depression is common low QoL and low QoL (together with depression)
among patients with ED with a frequency rang- predicts the development of ED.
ing from 8.7% to 43.1% [92], with an interrela-
tionship between depression and ED [92]. Cardiovascular Disease
Depression and diabetes share reciprocal suscep- There is enough evidence that ED shares the
tibility and comorbidities and the prevalence of same risk factors as cardiovascular diseases and
symptoms of depression in patients with diabetes in addition, ED in itself is actually considered an
is around 30% [93]. In this context, the effect of early sign of forthcoming cardiovascular disease
ED on mental health is of extreme relevance. In with a time window of 2–5 years before its devel-
the SUBITO-DE study, ED and severe depres- opment [97]. At least five meta-analyses have
sion were already associated in patients with confirmed the association between metabolic
newly diagnosed type 2 diabetes [83]. In Italian syndrome and its components with up to 2.6-fold
QuED study in type 2 diabetes, ED was associ- increase in ED, in particular fasting blood glu-
ated with higher diabetes-specific health distress, cose [98] and hypertension [99, 100]. Past and
worse psychological adaptation to diabetes, current smoking and cannabis use were associ-
severe depressive symptoms (present in 45.6% of ated with higher risk, while physical activity and
patients with frequent ED), lower scores in the moderate alcohol exerted a protective role in
mental components of QoL, and a less satisfac- other meta-analyses focused on these lifestyle
tory sex life [94]. The impact of ED on QoL was factors [97, 101]. Conversely, two large meta-­
independent of the presence of depression [94]. analyses showed that ED was associated with
After 3 years of follow-up, a worsening of physi- increased risk of cardiovascular disease, coro-
cal functioning scores was observed only in nary heart disease, stroke, and all-cause mortality
patients with ED at baseline, while the newly with the highest pooled relative risk of coronary
developed ED was preceded by a deterioration in heart disease (up to 1.62) [102, 103]. Moreover,
all QoL dimensions and a worsening in depres- the risk associated with ED was higher at younger
sive symptoms and was in turn associated with an ages, and a grading effect of ED severity in pre-
increase in depressive symptoms and decrease in dicting cardiovascular events was suggested
504 V. Spallone et al.

[103]. In the ED population not only conven- gonadism associated with both ED and cardio-
tional cardiovascular risk factors but also other vascular morbidity [97].
factors may increase the cardiovascular risk
including organic factors such as low testoster- Diabetic Neuropathy
one and PRL levels, psychological and relational Considering, among the many available studies,
aspects like depression and reduced sexual inter- those with larger sample sizes and standard
course [97]. assessment of CAN and ED, the overall findings
In summary, cardiovascular disease increases suggest that CAN is associated with ED [9, 81,
the risk of ED in diabetes, and in the general 109]. In a cross-sectional Italian study, peripheral
population there is evidence that (1) traditional vascular disease, cardiac disease, nephropathy,
cardiovascular risk factors increase the risk of autonomic neuropathy, polyneuropathy, diabetic
ED, (2) lifestyle changes are able to improve ED, foot, and retinopathy were all independent cor-
(3) ED is associated with a relative risk of devel- relates of ED in diabetes, with the greatest odds
oping cardiovascular disease ranging from 1.34 for autonomic neuropathy (4.3 vs. 3.6, 3.0 and
to 1.62 (a magnitude similar to that of main car- 2.5 of diabetic foot, polyneuropathy, and arteri-
diovascular risk factors), and (4) ED can be con- opathy) [81]. However, in a subsequent 2.8-year
sidered a low-cost biomarker for the need for follow-up study, the relative risk of ED associ-
more intensive cardiovascular risk factor ated with autonomic neuropathy was 1.16 com-
modification. pared to 3.79 of diabetic foot, 2.02 of coronary
artery disease, 1.97 of renal disease, 1.86 of poly-
Mechanisms Linking Erectile Dysfunction neuropathy, and 1.75 of peripheral vascular dis-
and Cardiovascular Risk ease [86]. In the UroEDIC study, ED development
The common risk factors, their mutual associa- at EDIC year 17 was associated with lower car-
tion, and the role of ED as a harbinger of cardio- diovascular reflex tests (CARTs) at DCCT close-
vascular events suggest a shared out and EDIC year 16/17 [9]. CAN diagnosis at
pathophysiological basis. The first link is vascu- EDIC year 16/17 was associated with 2.65 greater
lar disease: the artery size hypothesis suggests adjusted odds of ED or LUTS (not of ED only)
that an equally sized atherosclerotic plaque [9]. In the same cohort at EDIC year 17, CAN
would be tolerated less and compromise flow had unadjusted odds for ED of 2.82 [10].
more in the smaller penile arteries than in coro- Regarding diabetic polyneuropathy, the
nary arteries, which is the reason why ED will UroEDIC study showed an association between
precede by 2–5  years coronary artery disease confirmed diabetic polyneuropathy and ED
[104]. Another possibility regards an impairment [110]. Moreover, at EDIC year 17, the presence
of endothelium-dependent and endothelium-­ of neuropathic symptoms was associated with
independent vasodilation before the development unadjusted odds for ED of 2.15, while retinopa-
of atherosclerotic plaque and more likely to cause thy and microalbuminuria had odds of 2.11 and
ED than angina. The observed benefit of type 5 2.83, respectively [10]. In two small studies in
phosphodiesterase inhibitors (PDE5Is) on the patients with type 1 and type 2 diabetes, ED was
incidence of cardiovascular disease in men with associated with impairment in measures of both
ED [105] and of major adverse cardiac events in large (neuropathic symptoms, vibration percep-
men with diabetes and silent coronary artery dis- tion threshold, sural and peroneal nerve ampli-
ease [106] might involve endothelial dysfunc- tude, and conduction velocity) and small fiber
tion. In fact, PDE5Is in patients with diabetes or function (thermal thresholds, deep breathing test,
high cardiovascular risk were able to improve intraepidermal nerve fiber density, and corneal
penile and systemic endothelial function, reduce confocal microscopy parameters) [111, 112].
ET-1 and endothelial inflammatory mediators, These and other studies have advocated a greater
and increase circulating endothelial progenitor association of ED with abnormalities in small
cells [107, 108]. Another linking factor is hypo- than in large nerve fiber function [111–113].
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 505

In summary, CAN diagnosis in cross-sectional muscle cells are contracted. Sexual stimulation
studies is associated with ED with increased causes NO release from nervous terminations
odds up to 4, while in longitudinal studies its pre- (NANC neurons). The NO activates soluble gua-
dictive power is lower and equivalent or less than nylate cyclase raising intracellular cyclic guano-
that of other diabetic complications. Similarly, sine monophosphate (cGMP) and then
the presence of diabetic polyneuropathy is asso- intracellular Ca2+, resulting in a relaxation of the
ciated with at least double the risk of ED in both smooth muscle in the arteries and arterioles sup-
cross-sectional and longitudinal studies, with a plying the erectile tissue (Fig. 3). The process is
possible (but not definitively shown) preferential also enhanced by concomitant acetylcholine
link with small fiber function abnormalities. (Ach)-derived adenylate cyclase activation.
Increased blood influx fills the lacunar spaces of
 echanisms Linking Erectile Dysfunction
M corpora cavernosa, causing a compression of the
and Diabetic Neuropathy subtunical compressive venules, resulting in
The association of diabetic neuropathy with ED almost complete occlusion of venous outflow
has biological soundness. Impaired sensation of (venous-occlusion phenomenon). cGMP is
the penis takes away local information to the hydrolyzed by cellular phosphodiesterase-5
brain and the afferent arm of reflex erection, (PDE5), restoring smooth muscle contraction
abnormal motor function impacts on skeletal and allowing the penis to return to its flaccid
muscles participating in erection; autonomic neu- state. Detumescence is also mediated by adrener-
ropathy affects arterial dilation through impaired gic receptor reactivation, which leads to rhythmic
NO release and NO-synthase function of nonad- contractions of the cavernosal smooth muscle, a
renergic noncholinergic (NANC) neurons, and decrease in arterial diameter and thereby induc-
trabecular smooth muscle relaxation of the cor- ing venous outflow [118].
pus cavernosum [114]. In vitro examination of Testosterone plays a role in nearly every level
the corpus cavernosum tissue of men with diabe- of the erectile function, from central genesis of
tes showed impairment of autonomically medi- sexual desire to pelvic ganglions neurotransmis-
ated relaxation of smooth muscle as well as sion and smooth muscle and endothelial cells
reduction in endothelium-dependent relaxation function in the corpora cavernosa [119]. At the
[115]. Dysfunction and degeneration of the central level testosterone promotes the release of
NANC nerves of the corpus cavernosum contrib- stimulatory neurotransmitters such as dopamine,
ute to the NO defect, the main culprit of ED, oxytocin, and NO, with an influence during sex-
which follows also the loss of NO endothelial ual development and later in puberty and adult-
production/activity induced by many factors like hood mating behaviors. At the peripheral level,
the increase in AGEs formation, protein kinase C testosterone modulates structure, function, and
(PKC) activation, oxidative stress, arginase II, innervation of trabecular smooth muscle cells,
and RhoA/Rho-kinase activity (involved in penile the endothelial function of penile vessels, and
flaccidity), and the presence of androgen defi- fibro-elastic properties of the corpus cavernosum
ciency [116]. [119, 120].
ED can result from three main abnormalities:
(1) an inability to induce the erectile mechanism,
3.2 Physiology (2) a deficiency in the arterial blood supply of the
and Pathophysiology penis, and (3) insufficient maintenance of a
of Erectile Function proper lacunar blood concentration (venous-­
occlusive dysfunction). These conditions are not
The erectile mechanism is based on the mutual mutually exclusive, and different degrees of each
interaction of vascular, neural, hormonal, and can occur in the natural history of ED. While in
psychological processes [117]. The penis remains the past, in the general population under 40, ED
in its flaccid state as long as the vascular smooth was considered in most cases a purely psycho-
506 V. Spallone et al.

NITRIC OXIDE

PDE5i
Ca2+
Sequestration
5’ -GMP GC

GTP Phospholamban

PDE5 cGMP PKG IP3


Receptor

Cytoplasmatic Ca2+
MYPT reduction

MLKC

Fig. 3  The cyclic guanosine monophosphate (cGMP)- (MLKC), thereby inducing smooth muscle relaxation.
signaling pathway in smooth muscle cell in penile vascu- PKG promotes the lowering of intracellular calcium
lature. Nitric oxide activates guanylate cyclase (GC) through the inhibition of inositol trisphosphate (IP3)
causing increased synthesis of cGMP. cGMP binds to and receptor and phosphorylation of phospholamban in sarco-
activates cGMP-dependent protein kinase (PKG), thereby plasmic reticulum. cGMP is broken down at the catalytic
promoting a cascade of phosphorylation events resulting site of PDE5, PDE5Is have a high affinity for the PDE5
in the activation of myosin light chain phosphatase catalytic site, thereby blocking cGMP access to the site
(MLCP) that dephosphorylates the light chain of myosin and fostering cGMP accumulation

genic disorder, up to 70% of these cases can actu- reducing NO release from terminal nerves [118].
ally have organic etiologies [121]. Primary Radical pelvic surgery (nerve-sparing or non-­
psychogenic ED is dependent on the erectolytic nerve sparing) is still a common form of iatro-
(or anti-erectile) effect of noradrenaline neuro- genic ED [118]. Calcium channel blockers
transmission. Stressful or depressive events, gen- directly act on cavernosal ion channel flux, while
eralized anxiety disorders, or other β-blockers and thiazide diuretics exercise an indi-
psychological-impacting factors are generally rect reduction in arterial blood flow to the inter-
associated with this type of ED.  Organic ED nal pudendal artery. Finally, hypogonadism
often involves or determines a psychological represents a relevant cause of poor response to
component, affecting general mood, interper- first-line therapy of ED [97].
sonal relationships, and overall individual QoL
[122]. Vasculogenic is largely the most common
nonendocrine organic cause of ED, can involve 3.3 Multifactorial Pathogenesis
arterial inflow disorders and/or venous outflow of Diabetic Erectile
changes (venous-occlusion dysfunction) gener- Dysfunction
ally in the setting of generalized atherosclerosis
[97, 118]. Neurogenic causes derive from central In diabetes many pathogenetic factors of ED may
or autonomic neural damage, which converge in coexist and potentiate each other. Diabetes-­related
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 507

mechanisms and comorbidities—i.e., the compo- 3.3.1 Hypogonadotropic


nents of metabolic syndrome—can affect endo- Hypogonadism
thelial function, micro- and macrovascular Hypogonadotropic hypogonadism, character-
function, peripheral nerve function, smooth mus- ized by low serum testosterone, reduced sper-
cle contractility, psychological function, CNS matogenesis, and inappropriately low or normal
function, and androgen secretion (Fig.  4). The gonadotropins levels, is associated with obesity,
efficacy on ED of lifestyle interventions (based on metabolic syndrome, and diabetes [125]. The
Mediterranean diet, weight loss, and exercise) or prevalence of hypogonadism at the onset of dia-
statin treatment [123, 124] and, at least in type 1 betes is 17.2% [83] but can reach 50% in the
diabetes, intensive glycemic control [110] con- later stages [125]. This relationship is bidirec-
firms the pathogenetic role of mechanisms trig- tional and probably based on a vicious circle
gered by metabolic and glycemic changes. which involves either central or peripheral
Inflammation is gaining increasing pathogenetic mechanisms. Most probably, obesity represents
relevance also in female sexual dysfunction [124]. the major player. The increased aromatase activ-

Cofactors/comorbidities Diabetes Depression

Obesity
Hyperglycaemia
Oxidative
Hypertension stress Micro Neuropathy
Dyslipidemia angiopathy
Psychological
Physical inactivity
Inflammation Macro distress
angiopathy
Hypogonadism
mic

Endothelial dysfunction
otor
ono

orim
Aut

↓ Penile sensaon
Sens

↓ Cavernosal vasodilaon
↑ Cavernosal vasoconstricon
↓ Smooth muscle relaxaon

Erectile dysfunction
Fig. 4  Multifactorial pathogenesis of diabetic erectile leading to endothelial dysfunction and an impaired erec-
dysfunction. Cardiovascular risk factors and other meta- tion. Moreover, diabetic complications such as atheroscle-
bolic syndrome components exacerbate the effects of rotic disease, microangiopathy and neuropathy,
hyperglycemia-driven pathways such as AGEs formation diabetes-related hypogonadism, antihypertensive drugs
and PKC activation, thus promoting oxidative stress and (β-blockers apart from nebivolol, thiazide diuretics, and
low-grade inflammation. These changes impair the NO aldosterone antagonists), and psychological factors are
bioavailability and synthesis and favor an imbalance further pathogenetic factors
between vasodilating and vasoconstrictive mechanisms,
508 V. Spallone et al.

ity in adipose tissue enhances testosterone con- obtained as well as information on other, poten-
version into estradiol, leading to a suppression tially reversible, causes (smoking, drug, or
of central GnRH production [125]. Insulin resis- alcohol abuse). An objective score of erectile
­
tance reduces the central secretion of GnRH and function can be achieved through self-­
elevates inflammatory mediators [126] and administered validated questionnaires, which
these later may inhibit LHRH stimulated LH provide a measure of ED severity also useful for
release. Moreover, obesity related high leptin cardiovascular risk stratification. The short five-
concentrations play a role. Leptin has inhibitory question form of the International Index of
effects on Leydig cells, as suggested by human Erectile Function (IIEF-5) and the Sexual Health
models [127] as well as leptin resistance in the Inventory for Men (SHIM) are useful tools for
hypothalamic-­ pituitary axis reducing LH both diagnosis and assessment of treatment
release, as suggested in animal models [128]. response [133]. Nevertheless, structured inter-
Further, SHBG levels are reduced in men with views are generally considered a more reliable
obesity and/or type 2 diabetes and are nega- instrument than questionnaires as they tend to
tively associated with the risk of metabolic dis- build a stronger patient–physician relationship
orders [129]. Conversely, hypogonadism and reduce the risk of missing relevant informa-
increases lipoprotein lipase activity, free fatty tion. So far, the only validated interview on ED
acids uptake, and triglyceride content within that has proven useful in several clinical studies
adipocytes and stimulates adipocyte prolifera- is the Structured Interview on Erectile
tion and visceral adipose tissue deposition, con- Dysfunction (SIEDY), composed of 13 items
tributing to visceral obesity [125]. Low exploring male sexual function [134]. A psycho-
testosterone levels are associated with an logical assessment is recommended in patients
adverse metabolic profile, promote insulin resis- with psychiatric disorders or young men with
tance in men by impairing mitochondrial func- short diabetes duration. It should be taken into
tion [125], and may impair endothelial integrity account that psychogenic aspects are present in
and promote inflammation [97]. different degrees in all patients independently of
the disease stage.

3.4 Diagnosis of Diabetic Erectile 3.4.2 Physical Examination


Dysfunction An accurate physical examination in addition to
heart rate, BP, waist circumference, and BMI
The diagnostic pathway of ED does not differ should be focused on the general body habitus,
between diabetes and the general population including the chest (body hair distribution and
[130, 131]. presence of gynecomastia) and genitalia (testic-
ular size, structure, consistency and secondary
3.4.1 History and Questionnaires sexual habits), seeking for signs of hypogonad-
A detailed family, medical and sexual history, is ism or other comorbidities [135]. The cremas-
the primary approach, including the timing of teric reflex can also be evaluated. A normal reflex
onset, severity, duration of ED, presence of noc- can be elicited if the thoracolumbar erection cen-
turnal erections, and impact on the patient’s sex- ter is intact. Evaluation of the penis needs to
ual and general QoL [132]. A complete exclude Peyronie’s disease, phimosis or frenu-
medication list, including supplements, should be lum breve, balanitis, and other diabetes-related
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 509

conditions affecting erectile response. In selected mation on penile hemodynamics to distinguish


high-­risk patients or patients with LUTS, a digi- arterial insufficiency and venous-occlusive dys-
tal rectal examination and prostate-specific anti- function from other causes of ED and improve
gen test are recommended [136]. Neurological cardiovascular risk stratification [141].
evaluation in patients with diabetes is appropri- Moreover, it is useful in patients with ED who
ate for frequent and early coexistence of diabetic failed to respond to oral agents. Peak systolic
neuropathy: clinical diagnosis includes assess- velocities after PGE-1 injection >35  cm/s are
ment of neuropathic and autonomic symptoms considered normal and values <25 cm/s sugges-
and signs using easy-to-use questionnaires and tive of arterial ­insufficiency (92% of accuracy).
hand-­held devices, while CARTs allow a con- Cavernous venous-­ occlusive dysfunction is
firmed diagnosis of CAN [60, 137, 138]. Vice demonstrated on Doppler ultrasounds by a
versa annual screening for erectile dysfunction rapid detumescence despite normal peak sys-
in men with other forms of diabetic neuropathy tolic and end-diastolic velocities (>5  cm/s).
is recommended by guidelines [60] and is also Vascular resistive index can be assessed as the
advisable in any patient with type 2 diabetes or difference between peak systolic and end-dia-
long-­standing type 1 diabetes. stolic velocities divided by peak systolic veloc-
ity (normal values <0.75) [142]. Penile
3.4.3 Laboratory Investigations arteriography is reserved to young men with
They are driven by the patient’s history and arterial trauma and abnormal duplex hemody-
physical examination findings and include gly- namics or to identify candidates for vascular
cemic control, lipids profile, renal and hepatic reconstructive surgery.
function for cardiovascular and metabolic risk Generally, most men with ED can be managed
stratification [131]. Morning fasting total testos- in the primary care setting. In the general popula-
terone assay and SHBG to calculate free testos- tion, ED requires further cardiovascular workup
terone are always indicated in men with diabetes according to the Princeton III Consensus Panel
and ED as well as in men with ED and signs of [131], and in patients with diabetes tight control
hypogonadism, hypoactive sexual desire, or of cardiovascular risk factors should also be
incomplete response to PDE5Is [139]. If total considered.
testosterone results <12 nmol/L (<346 ng/dL), a In summary, a cost-effective diagnostic
second-morning measurement after at least a approach to patients with ED (Fig. 5) relies on
week, together with serum LH and PRL levels, a careful medical history and physical exami-
is required in order to both confirm hypogonad- nation. Routine laboratory testing is limited
ism and determine its etiology (central or and aimed at a proper cardiovascular and
peripheral) [139]. metabolic risk stratification. Considering the
relationship between diabetes and hypogonad-
3.4.4 Instrumental Procedures ism, serum testosterone levels, SHBG, LH,
A role for rigidity testing with RigiScan is still PRL are more important steps than in men
useful for medical-legal cases and in investiga- without diabetes. The vascular investigation
tive pharmacological studies to assess treat- with penile duplex Doppler ultrasound might
ment impact [140]. Penile duplex Doppler be more appropriate in patients with diabetes,
ultrasound before and after an intracorporal especially for those with high cardiovascular
injection of a vasoactive drug provides infor- risk.
510 V. Spallone et al.

Diagnosis Medications list and psychological assessment


Medical and sexual history
1st Step Validated self-questionnaires (e.g., IIEF-5, SHIM, SIEDY)

General habitus, genitalia, signs of hypogonadism,


Physical examination
anthropometric measures, BP, neurological examination*

Metabolic, liver and kidney markers, hormonal status


2nd Step Laboratory tests
Assess global cardiovascular risk, exclude hypogonadism

Selected Penile duplex doppler ultrasound before and after an


3rd Step Instrumental procedures
patients intracorporal injection of a vasoactive drug

Treatment
General Measures Lifestyle interventions Physical activity, diet, weight loss

Glucometabolic control Optimization of metabolic indices and comorbidities

Specific Measures
Psychosexual strategies Psychological counseling, partner involvement
1st Step
Oral treatment with PDE5i
1st line
In non-responders, switch to another PDE5i, dose escalation
or chronic daily treatment with tadalafil
Pharmacological strategies 2nd line
IUS or ICI (e.g., PGE1)
Hypogonadism
TRT in monotherapy or in combination

Selected
2nd Step Mechanical devices External vacuum devices
patients
Selected Penile prosthesis
Surgical Interventions
patients Vascular reconstructive surgery

Fig. 5  An algorithm for a diagnostic and therapeutic of inefficacy, in hypogonadal men, TRT should be started,
approach to diabetic erectile dysfunction. A preliminary while in eugonadal men, escalation of the PDE5I dose, a
assessment is based on medical history, validated self-­ change of PDE5I, the use of daily dosing tadalafil could
questionnaires, and physical examination. A second step be attempted. Psychological counseling is useful for an
with routine laboratory testing for a proper cardiovascular underlying psychosexual dysfunction (including the part-
and metabolic risk stratification. Hormonal assessment is ner). IUS or ICI should be attempted in second-line,
an important diagnostic step. The vascular investigation, whereas external vacuum device and surgery remain lim-
like penile duplex Doppler ultrasound, is limited to ited to a small number of selected individuals [139, 143–
selected patients [130, 131]. Therapeutic management is 146]. PDE5Is type 5 phosphodiesterase inhibitors, TRT
based on lifestyle changes and the optimization of gluco- testosterone replacement therapy, IUS intraurethral
metabolic control and comorbidities. The first-line oral alprostadil suppositories, ICI intracavernosal injections
therapy with PDE5Is is the treatment of choice. In event

3.5 Treatment of Diabetic Erectile AHEAD study showed that weight loss inter-
Dysfunction vention was mildly helpful in maintaining erec-
tile function in older overweight/obese men
The stepwise management of ED includes gen- with type 2 diabetes [149], while intensive gly-
eral and specific measures. cemic control was able to reduce the incidence
of ED in the DCCT/EDIC study in type 1 diabe-
3.5.1 Lifestyle Interventions tes [110].
Lifestyle modifications target ED and cardio-
vascular risk by promoting physical activity 3.5.2 Psychosexual Strategies
[147], weight loss, Mediterranean diet [148], In patients with an evident psychological involve-
avoidance of smoking, drugs and alcohol abuse, ment, as is possible with young men with new-­
and glycemic and lipidic control. The Look onset diabetes, psychosexual counseling should
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 511

be encouraged besides pharmacological therapy. hypogonadal men [107]. Moreover, the beneficial
The counseling should also be extended to the effects of PDE5Is in patients with diabetes could
partner, as it has shown better outcomes in resolv- go beyond the treatment of ED with cardioprotec-
ing relationship conflicts and improving sexual tive effects, an improvement in markers of dia-
function [150]. betic cardiomyopathy, and endothelial dysfunction
[107, 155]. The side effects of PDE5Is are usually
3.5.3 Pharmacological Treatment: mild and occur in less than 40% of patients like
PDE5Is headache, nasal congestion or facial flushing,
Oral PDE5Is are effective, safe, and well-­tolerated nausea, and mild reversible visual disturbances.
therapies for the treatment of men with ED and Although without a definitely proven association,
are for most men first-line treatment [143]. deafness and non-arteritic ischemic optic neurop-
Sildenafil, first in class, and later vardenafil, athy are uncommon adverse events of PGE5Is use
tadalafil, avanafil, and other available PDE5Is requiring urgent medical assessment and with-
have shown efficacy in a wide range of etiologies, drawal [156, 157]. Some caution should be used
including cardiovascular diseases and diabetes, with concomitant antihypertensives drugs and
with an overall success rate of up to 76% depend- α-blockers, whereas co-administration with
ing on study populations [143, 151]. Their com- nitrates is prohibited [158]. In the event of initial
petitive inhibition of PDE5 increases cGMP unresponsiveness to the treatment, switching to at
concentrations after NO release. Consequently, least one other PDE5I class member should be
PDE5Is cannot induce an erection without proper attempted as up to 50% of patients has shown to
sexual stimulation. An adequate time before sex- be respondent [159].
ual intercourse is also required to allow peak
absorption of the drugs. Patients should be 3.5.4 Pharmacological Treatment:
instructed on optimal conditions for effective Testosterone
medications function. Sildenafil and vardenafil Men with ED and hypogonadism often fail to
reach a median Cmax (maximum observed achieve initial clinical success with PDE5Is and
plasma concentration) values 1 h after administra- thus testosterone replacement therapy (TRT) is
tion with a serum half-life of 3–5 h. Both drugs indicated. Clinical trials with TRT in men with
showed better efficacy if taken with an empty type 2 diabetes and hypogonadism documented
stomach because lipids in foods decrease and an improvement in sexual function and erection,
delay absorption. Food impact on pharmacoki- beneficial effects on various aspects of sexual
netic properties is less evident with tadalafil and function like sexual desire, a possible delay of
avanafil [152] (Table 1). A network meta-analysis some weeks for a benefit in erectile function
of 15 studies confirmed that PDE5Is are effective, [160], and a lower efficacy compared to men
safe, and well tolerated in men with diabetes and without diabetes. A combination of TRT and a
ED, found better mean differences in the domain PDE5I can further improve erectogenic outcomes
of erectile function of IIEF compared to placebo [161]. TRT requires monitoring of prostate-­
for vardenafil, mirodenafil, and sildenafil, and specific antigen, hematocrit, and cardiovascular
suggested an overall possible superiority of varde- performances [162]. A meta-analysis of six stud-
nafil and mirodenafil, while no significant differ- ies in men with type 2 diabetes with/without
ences between regular and on-demand regimens hypogonadism showed a moderate improvement
of PDE5Is were observed [154]. In a previous of sexual desire and erectile function with TRT
clinical trial of 54 men with type 2 diabetes a but emphasized the lack of information on the
6-month 20  mg daily treatment with vardenafil long-term risks of treatment and the need for a
was able to improve erectile function and in addi- preliminary risk/benefit assessment [144]. An
tion serum IL-6 levels and serum testosterone, improvement in metabolic parameters with TRT
which returned within the eugonadal range in 13 has been also documented, mostly in patients
512 V. Spallone et al.

with type 2 diabetes [125], with less data in type 3.5.6 Mechanical Devices
1 diabetes [163], but there is not definitive evi- Vacuum erectile devices are long-term, inexpen-
dence on TRT’s beneficial effects on hypogonad- sive non-surgical tools for patients with ED refrac-
otropic hypogonadism-related metabolic tory to first-line therapy. They consist of a
features. cylindrical mechanical pump that creates negative
pressure that artificially draws blood into the penis.
3.5.5 Pharmacological Treatment: Erection is maintained by an elastic band placed
Intracavernosal around the base of the penis but application should
and Intraurethral Therapy not exceed 30 min. Even though the efficacy rate is
Second-line treatment for PDE5Is-non-­responders reportedly high (above 70%), the long-term drop-
includes intraurethral vasoactive PGE1 (alprosta- out rates are also high. Petechiae and hematomas
dil) suppositories (IUS) or intracavernosal injec- are the most common mild adverse events and can
tions (ICI) with vasoactive drugs (Table  1). cause discontinuation [138]. Caution is needed
Intraurethral alprostadil absorption causes an when taking anticoagulants [167].
immediate increase in intracellular cyclic AMP,
smooth muscle cells relaxation in the corpora cav- 3.5.7 Surgery
ernosa and a consequent erection. IUS efficacy has Surgical interventions are indicated for patients who
been described in up to 56% of cases, with patients are refractory to first- and second-line treatments, or
with organic ED responding better, including with contraindications to other treatments, or with
those with diabetes [164]. Dosages, lag time, and adverse reactions or acquired neural or vascular
common side effects are described in Table  1 penile dysfunctions after pelvic trauma. Penile
[145]. ICIs use vasoactive substances self-injected implants prostheses divide into two main types: mal-
directly into the corpora cavernosa via a small leable and inflatable. Both are surgically inserted into
needle to obtain an immediate erection. Alprostadil the corpora cavernosa in order to supply a physiolog-
ICI is the single agent most widely used, with a ical erection. The surgical approach can be infra-
good efficacy rate and fewer systemic side effects pubic, penoscrotal, or subcoronal [146]. Malleable
than other agents [145]. The erection occurs after prostheses rely on the patient’s physical manipulation
10 min and may be enhanced by sexual stimula- for changing from a straight to a bent position.
tion. In an open-label study in patients with diabe- Although easy to handle, they often can cause social
tes a satisfactory erectile response was achieved embarrassment and discomfort due to their mechani-
after 99% of injections without differences cal rigidity. The two-piece inflatable prosthesis con-
between type 1 and type 2 diabetes and with a fair sists of two cylinders inserted in the corpora cavernosa
safety profile [165]. Other vasoactive medications and a small scrotal pump, which drives fluid move-
for ICI include papaverine, phentolamine, and ment from a small reservoir inside the patient’s lower
atropine for both mono- or multiple combinations abdomen when an erection is desired. Although it
and possible dose titration. Priapism is a major provides similar rigidity to a malleable device, it also
problem with IUS and ICI, and patients should be enables better flaccidity when deflated. Skin erosion,
informed to require urgent medical intervention if fluid leakage, genitourinary infections, and the pros-
the condition emerges. Fibrosis, pain, bruising, thesis’s mechanical failure are the most documented
patient’s reluctance to penile injections, and costs complications, albeit reduced in frequency with the
related drop-out are other important issues to be latest devices [168].
considered [145] (Table 1). Training in self-injec- Penile revascularization surgery is currently
tion technique as well as counseling might favor indicated for young patients (<55  years of age)
treatment adherence. Combined therapy with ICIs with penile vascular Doppler ultrasound demon-
and PDE5Is can also be effective in the event of strated and isolated injuries. A microvascular
single-­class failures [166]. anastomosis between the inferior epigastric
Table 1  Comparison of the main oral type 5 phosphodiesterase inhibitors (PDE5Is) and intraurethral vasoactive PGE1 suppositories (IUS) or intracavernosal injections (ICI)
with vasoactive drugs [108, 143, 153]
PDE5i compound Dose range Tmaxa (h) T1/2a IC50 for Main adverse events Comment
(h) PDE5a (%)
(nmol/L)
Sildenafil 50–100 mg 0.95 3.98 3.7 Headache (19%), First in class; delayed
flushing (14%), absorption after high fat
dyspepsia (9%) meals
Tadalafil 10–20 mg 2 17.5 1.8 Headache (21%), Efficacy for up to 36 h;
5 mg daily dyspepsia (17%), back efficacy not reduced by high
pain (6%) fat meals
Vardenafil 10–20 mg 0.66 3.9 0.091 Headache (16%), Delayed absorption after
flushing (12%), nasal high fat meals
congestion (10%)
Avanafil 5–200 mg 0.75 5.1 5.2 Headache (7–8%), Highly selective; efficacy
flushing (3–5%) not reduced by high fat
meals
IUS or ICI compound Administration Dose range Time to onset Main adverse events Comment
(%)
Alprostadil IUS 125–1000 μg 10 min Pain (30%), urethral Easy and available;
bleeding (5%), priapism uncommon systemic side
(<1%) effects
Alprostadil ICI 5–20 μg 5–15 min Pain (25%), priapism Most used; uncommon
(<2%), fibrosis (<5%) systemic side effects
Papaverine + phentolamine (Bimix) ICI 30 mg/mL + 0.5 mg/ 5–10 min Pain (14%), priapism Not licensed for ED
mL (5%), fibrosis (13%) treatment; only available as
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes

galenic preparation
Papaverine + phentolamine + alprostadil ICI 30 mg/mL + 1 mg/ 5–10 min Pain (15%), priapism Not licensed for ED
(Trimix) mL + 10 μg/mL (3%), fibrosis (<5%) treatment; only available as
galenic preparation
VIP + Phentolamine ICI 25 μg + 1–2 mg Not recorded Flushing, headache, Approved in Denmark, the
priapism (0.06%) UK, and in New Zealand
a
Referred to fasting state, higher recommended dose. Tmax: time-to-maximum plasma concentration; T1/2: plasma elimination halftime; Cmax: maximum plasma concentration;
IC50: half maximal inhibitory concentration; VIP: Vasoactive intestinal polypeptide
513
514 V. Spallone et al.

artery and the penile dorsal artery is performed to inflammatory properties of GLP-1RA and DPP-4i
ensure arterial blood supply to corpora cavernosa [153] or on DPP-4i ability to promote vascular
[169]. Due to high procedure related risks and repair and endothelial function [173], while a
often limited long-term results [170] penile vas- 4-week treatment with empagliflozin increased
cular surgery has little application in diabetes. nitrergic relaxation of erectile tissues in a type 2
diabetes animal model [174]. From a clinical
3.5.8 Future Treatments standpoint, in a 12-week observational study a
Future therapies should address the need of patients combined treatment with exenatide and metfor-
nonresponders to first-line PDE5 inhibition. Low- min induced a greater improvement in total tes-
intensity extracorporeal shockwave therapy has tosterone levels and sexual function than
preliminarily shown some efficacy. Its proposed glimepiride-metformin combination in obese men
action mechanism ranges from improvement in with type 2 diabetes [175], with, as expectedly, an
cavernous hemodynamics and increased angio- increase in testosterone and sexual function being
genic factors to restoration of smooth muscle cell strongly related to weight and waist circumfer-
activities [171]. Biotherapies to regenerate cavern- ence loss, which are the known effects of exena-
ous endothelial cells and nerves and to inhibit cav- tide treatment. The two- to threefold increase in
ernous fibrosis have also been investigated using risk of genital infections associated with SGLT2i
growth factors, gene or protein therapy, and human should be taken into account for appropriate pre-
umbilical cord stem cells [167], with the latter vention and timely treatment [176].
showing beneficial effects on erectile function in a In summary, very few animal studies and
few men with severe type 2 diabetes when admin- observational clinical studies have investigated
istered locally [172]. Phase II–III studies are the impact of DPP-4i, GLP-1RA, and SGLT-2i
required for these regenerative therapies. on erectile and sexual function. The limited
In summary, a therapeutic algorithm for ED in data with GLP-1RA in different subsets of
men with diabetes (Fig. 5) is based on patient’s patients do not provide comparable results
clinical and laboratory profile. First-line oral [77]. Nevertheless, novel drugs have shown
therapy with PDE5i is the treatment of choice, promising metabolic effects and weight loss
but patients with diabetes could be non-­ might be a way, albeit indirect, to improve
responders. In this case, in hypogonadal men, gonadal and sexual function in patients with
TRT should be started, while in eugonadal men, diabetes and sexual dysfunction [77].
an escalation in the PDE5i dose, a change of
PDE5i, or the use of daily dosing tadalafil could
be attempted. Psychological counseling to 4 Ejaculation Dysfunction
improve patients’ sexual health and overall QoL in Diabetes
is a useful tool for an underlying psychosexual
dysfunction and should include the partner. More 4.1 Epidemiology of Ejaculation
invasive treatments, like IUS and ICI, should be and Orgasmic Dysfunction
chosen in second-line, whereas vacuum device in Diabetes
and surgical interventions remain often limited to
a small number of selected individuals. Premature ejaculation (PE) is defined according
to the International Society for Sexual Medicine
[177] as characterized by ejaculation that nearly
3.6 Novel Antihyperglycemic always occurs prior to or within about 1 min of
Drugs and Male Sexual vaginal penetration from the first sexual experi-
Function ence (lifelong PE), or a clinically significant
reduction in latency time, often to about 3 min or
Indirect evidence from animal models suggests a less (acquired PE); the inability to delay ejacula-
possible benefit of the latest antihyperglycemic tion in nearly all vaginal penetrations; and nega-
drugs based on anti-atherogenic and anti-­ tive personal consequences and/or the avoidance
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 515

of sexual intimacy [78, 177]. In real-life clinical 26.1% and 37.6% of participants, aged
practice, the same definition could be applied to 57–84 years, with known diabetes referred inabil-
other sexual stimuli and non-heterosexual set- ity to climax and a lack of interest in sex com-
tings [178]. Prevalence of PE in the general pop- pared to 15.9% and 24.8% of those without
ulation ranges from 8% to 30% [179]. A few diabetes, respectively [184]. In the SUBITO-DE
studies described an association between PE and study, hypoactive sexual desire was present in
diabetes with a prevalence up to 40.2% [180] and 58.4% [83]. In the UroEDIC study, low sexual
of 28.3% in the SUBITO-DE study in men with desire and orgasmic dysfunction were present in
recently diagnosed type 2 diabetes [83]. In young 40% and 14%, respectively, and disorders of
men with type 1 diabetes, the prevalence of PE orgasm, including retrograde ejaculation and
was 24% and both HbA1c and glycemic excur- anejaculation closely overlapped with ED [10].
sion in the hypoglycemic range were positively In the same cohort, men reporting only orgasmic
related to PE severity [181]. ED and PE can co-­ dysfunction presented a predominantly psycho-
occur in the same subjects in up to 50% of cases, genic phenotype, while men with orgasmic dys-
thus creating a vicious cycle [178] in both the function and concomitant ED showed a mixed
general and diabetic population with PE being phenotype with a physiological predominance
associated with an odds ratio for ED of 3.68 and and more severe diabetic and metabolic altera-
4.41, respectively [83, 180]. tions [185].
There is a paucity of reliable prevalence data In summary, albeit with evidence based on
on delayed ejaculation (defined as a distressing only a few studies with an inhomogeneous defini-
lengthening of ejaculatory latency) with rates tion of conditions, ejaculation and orgasmic dys-
varying from 1% to 10% in the general popula- function appear to be more prevalent in diabetes
tion [179, 182]. In SUBITO-DE study, delayed than in the general population, with rates from
ejaculation was present in 32.9% [83]. 24% to 40% for PE, of 33% for delayed ejacula-
Retrograde ejaculation is defined as the expul- tion, up to 35% for retrograde ejaculation, from
sion of seminal fluid into the bladder because of 25% to 58% for low sex desire/interest, and from
bladder neck dysfunction in the presence of oth- 14% to 26% for orgasmic dysfunction. There is
erwise normal emission and expulsion [179]. close overlapping between ejaculation dysfunc-
Prevalence of retrograde ejaculation ranges from tion, orgasmic dysfunction, and ED, suggesting
0.3% to 2% in patients attending fertility clinics. common and interactive pathogenesis and the
Given the main autonomic control of anterograde need for screening for the other two conditions in
ejaculation, as expected, diabetic autonomic neu- patients referred for one of them. Furthermore,
ropathy was found to be associated with retro- ejaculation dysfunction might also impair
grade ejaculation in case reports [182]. True fertility.
prevalence might be higher than actually docu-
mented, as suggested by a small study where
34.6% of men with diabetes had retrograde ejac- 4.2 Premature Ejaculation
ulation compared to 0% of control subjects [182]. in Diabetes
Ejaculatory dysfunction was the most common
cause of infertility among patients with diabetes 4.2.1 Pathophysiology of Premature
attending an infertility clinic [183]. Ejaculation
The prevalence of decreased sexual interest or Ejaculation depends on the integrity of the auto-
desire in the general population ranges from 15% nomic nervous system and its central and periph-
to 25% until 60 years of age, after which preva- eral neurotransmitters [177]. It consists of two
lence sharply increases [179]. The prevalence of main phases—i.e., emission and expulsion—and
orgasmic dysfunction is difficult to assess and orgasm is generally associated with ejaculation
ranges from 1.8% to 19.4% with most values although the two processes are physiologically
being ≥10% [179]. In a population-based study, different [186]. PE has been associated with
516 V. Spallone et al.

hypersensitivity of the glans penis, abnormali- between dapoxetine and lidocaine/prilocaine,


ties of serotonergic neurotransmission, hyper- PDE5i, and other SSRIs, after screening for
thyroidism, chronic prostatitis, diabetes, high depression. Their off-label use requires patient’s
testosterone and low prolactin levels, sexual informed consent [178]. Psychological/behav-
desire disorder, and ED [177, 178]. Moreover, ioral interventions are indicated to manage the
sexual performance anxiety and psychological psychosocial aspects of PE, and a pharmacologi-
or relationship problems often coexist with PE cal and psychological combined approach is sug-
as either a cause or a consequence of PE [187]. gested [178].
Such comorbidities should be considered as pos-
sible causes of acquired PE and worsening fac-
tors of lifelong PE for which a genetic 4.3 Retrograde Ejaculation
pathogenesis has been hypothesized with incon- in Diabetes
clusive evidence [177]. Regarding a PE and dia-
betes association, possible underlying 4.3.1 Pathophysiology of Retrograde
mechanisms might involve neurologic, neu- Ejaculation
rotransmitter, and psychologic changes without Any factor that invalidates bladder neck contrac-
any supporting findings apart from changes in tion may lead to retrograde ejaculation. These
metabolism of NO that in male rats inhibits sem- factors can be mechanical, from anatomical dis-
inal emission, probably by decreasing sympa- ruption during transurethral prostate resection,
thetic nervous system activity [188]. pharmacological, due to α-blockers use, or neu-
rological, from impaired sympathetic control of
4.2.2 Management of Premature the internal vesical sphincter [189] as can occur
Ejaculation due to diabetic autonomic neuropathy in long-­
Medical and psycho-sexological history should standing diabetes.
establish the PE diagnosis, differentiate lifelong
PE and acquired PE, and assess the erectile func- 4.3.2 Management of Retrograde
tion and the impact on relationships and Ejaculation
QoL. Partner and patient self-estimation of ejacu- Retrograde ejaculation should be suspected in any
latory latency are currently the preferred assess- patient with aspermia or hypospermia and diag-
ment of PE as a proxy measure of objective nosis is based on the demonstration of 10–15
intravaginal ejaculatory latency time assessment, sperm per high powered field in post-ejaculation
whereas stopwatch measures have been consid- urine [190]. In the case of pharmacological form,
ered disruptive of sexual spontaneity [177]. In the offending drug should be, if possible, stopped.
this context, questionnaires such as the Premature Medical treatments include α-agonists such as
Ejaculation Profile (PEP) or the Index of ephedrine, pseudoephedrine, and phenylpropa-
Premature Ejaculation (IPE) are useful tools nolamine, and imipramine, a tricyclic antidepres-
[177]. A physical examination is highly advisable sant with α-adrenergic action, used alone or in
and associated/causal diseases such as ED, thy- combination with an α-agonist. Their efficacy has
roid dysfunction, or prostatitis should be assessed. also been demonstrated in patients with diabetes
The only currently on-label pharmacotherapy [191]. Moreover, endourethral injection of colla-
for both lifelong PE and acquired PE is oral gen type 2 showed promising results in patients
dapoxetine, a short-acting selective serotonin with type 1 diabetes [192]. In a patient wishing to
reuptake inhibitor (SSRI). Local anesthetic (lido- conceive and refractory to drugs, post-­ejaculation
caine/prilocaine) spray represents the second bladder harvest of sperm or testicular sperm
officially approved drug for this indication [189]. extraction can allow for the performance of medi-
Off-label therapies include a combination cally assisted reproductive techniques [189].
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 517

5 Female Sexual Dysfunction 68.6% in women with type 2 diabetes, ranging


in Diabetes from 17% in Italy to 94.4% in Iran [195]. In the
UroEDIC I in sexually active women with type 1
5.1 Definition and Epidemiology diabetes, FSD was reported by 35.4%, including
of Diabetic Female Sexual decreased desire, problems with orgasm, lubrica-
Dysfunction tion, and arousal, pain, and low overall sexual
satisfaction by 25% [196].
Female sexual dysfunction (FSD) is a complex BMI was the only factor associated with FSD
condition affecting the physical and emotional in the cited meta-analysis in both types of diabe-
well-being of women of all ages and ethnicities. tes [194], while depression and marital status
According to the Diagnostic and Statistical were the only independent predictors in type 1
Manual of Mental Disorders (DSM)-5, FSD diabetes in the UroEDIC I study, with odds ratios
includes four primary diagnoses: female sexual of 2.08 and 2.49, respectively [196]. Depression
interest and arousal disorder, female orgasmic was also independently associated with the
disorder, genitopelvic pain, and penetration dis- Female Sexual Function Index (FSFI) score in
order. In contrast, the International Classification both type 1 [197] and type 2 diabetes [198]. In
of Diseases and Statistics (ICD)-10 distinguishes young women with type 1 diabetes those on insu-
between nonorganic and organic sexual dysfunc- lin pumps had a prevalence of FSD equal to con-
tions. This approach did not consider the multi- trol women and lower than women on multiple
factorial etiology of FSD and has been surpassed daily injections [197]. In the UroEDIC II study,
by the newest ICD-11 classification that includes at EDIC year 17, CAN and CARTs were inde-
both the components formerly classified in men- pendently associated with FSD (present in 41%
tal and behavioral disorders and those located of sexually active women) and predicted the sub-
among genitourinary system diseases [193]. sequent development of FSD [20], with the diag-
Regardless of several methodological discrep- nosis of CAN having an odds ratio of 1.67 for
ancies and an inconsistency in epidemiological FSD [10]. In the UroEDIC II study, among
data, the prevalence of women in the general women with FSD, 52% showed FSD in isolation
population who report at least one sexual dys- and 48% in association with one or more among
function among desire, arousal, orgasmic, and urinary incontinence, LUTS, and urinary tract
genitopelvic pain dysfunction is around 40–50%, infections [10]. This co-occurrence might docu-
irrespective of age [179]. Risk factors are aging, ment both shared pathogenesis and mutual inter-
diabetes, hypertension, genitourinary disease, action between diabetes, urinary, and sexual
psychiatric/psychological disorders, cardiovas- function also in women.
cular disease, cancer, and other chronic diseases. In summary, epidemiological data on FSD
Sociocultural factors include social isolation, are limited and of poor consistency in the gen-
financial and employment status, religious eral and diabetic population. Women with dia-
beliefs, education level, and physical activity betes are less sexually active than those without
[148]. The NSHAP study indicated that 33% of diabetes. FSD appears to be more common in
women aged 57–85  years with diabetes were women with diabetes with even doubled the
sexually active compared to 45.5% of women associated risk. Psychosocial aspects such as
without diabetes [184]. A meta-analysis of 26 depression were the main clinical correlates,
studies showed that diabetes was associated with especially in type 1 diabetes, with additional
an odds ratio for FSD of 2.27 in type 1 and 2.49 in variables as BMI mainly in type 2 diabetes. A
type 2 diabetes [194]. A more recent meta-­ predictive value for FSD was shown for CAN in
analysis showed an overall prevalence of FSD of DCCT/EDIC study.
518 V. Spallone et al.

5.2 Physiology FSD were lacking until the DCCT/EDIC study


and Pathophysiology that reported an association between CAN and
of Female Sexual Function FSD [10, 20]. Finally, diabetes-related modifica-
tions in steroid hormones and comorbidities,
The physiology of female sexual desire and such as polycystic ovarian syndrome and thyroid
arousal involves a combination of psychological, diseases might intervene in FSD [202].
environmental, nervous, and hormonal factors.
As a consequence of sexual arousal and erotic
stimulation, vasodilators, such as NO and vasoin- 5.4 Diagnosis of Diabetic Female
testinal polypeptide, are released, thank to both Sexual Dysfunction
parasympathetic and sympathetic nerves, leading
to vaginal vasocongestion, smooth muscle relax- Assessing FSD can be very challenging given the
ation, and arteriolar dilation, which increases multiple involved factors to be examined. The
transudation of interstitial fluid, promoting lubri- International Society for the Study of Women’s
cation [199]. Estrogens are involved in increasing Sexual Health has proposed a care process with
vasoprotective and vasodilatory effects through- practical recommendations to screen for and
out the nervous system [200]. Similarly, andro- detect sexual problems in women, focusing on
gens play a role, despite the lack of relationships patient-centered communication [193].
between any FSD and having a low serum testos-
terone or androstenedione level in a large 5.4.1 Questionnaires and Clinical
Australian study [201]. Assessment
The FSFI questionnaire, one of the most used,
includes 19 items in six domains (desire, arousal,
5.3 Pathogenesis of Diabetic lubrication, orgasm, satisfaction, and pain)
Female Sexual Dysfunction addressing sexual function over 4 weeks: a total
score ≤26 identifies a risk of FSD [193]. A vali-
Although FSD appears to be mainly related to dated abridged FSFI version, the FSFI-6, includes
social and psychological factors, like depression six questions and takes 3 min, with a score <19
[196, 198], diabetes can affect the mechanisms of requiring further investigations [204]. Once a
female sexual function at many levels. Animal FSD has been detected, a clinical assessment
models documented diabetes-related changes in based on general medical history (including med-
contractile and relaxant capacity of vaginal mus- ications) and a physical examination, searching
culature, reduction in clitoral and vaginal blood for contributing factors (infectious, inflamma-
flow, and fibrosis development [202]. tory, neoplastic, neurologic) and vaginal atrophy,
Theoretically, diabetes-related endothelial skin lesions, or pain, is essential [193]. A com-
­dysfunction might cause vascular insufficiency in plete diabetes history should be collected. The
genital tissues. Higher clitoral resistance to blood patient’s psychological state should also be
flow was associated with both metabolic syn- investigated, as this is a key player in the whole
drome and impaired sexual arousal [202, 203]. process of her sexual enjoyment [193].
Although FSD is more common in metabolic
syndrome and also linked with inflammation 5.4.2 Laboratory Testing
markers, there is no definitive evidence of an This may be helpful (e.g., vaginal pH) but usually
association between cardiovascular risk factors unnecessary. Testing for sexually transmitted
and FSD nor the prognostic cardiovascular value infections and other vaginal pathogens may be of
of FSD [148, 200]. Moreover, given the neuro- help in presence of pain. Other laboratory tests,
regulation of female sexual function, the expected such as TSH and PRL levels, may be indicated
data of a role of diabetic neuropathy in diabetic based on symptoms or signs [193].
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 519

5.4.3 Instrumental Procedures showed lower clitoral peak systolic velocity and
Somatosensory evoked potentials and clitoral end-diastolic velocity and higher resistance index
electromyography could be used to assess the compared with healthy controls [206].
muscular and neural system [205]. Different Furthermore, the clitoral pulsatility index was
diagnostic systems evaluate female genital blood correlated with metabolic syndrome and with a
flow, whose increase is the main marker of sexual subjective decrease in sexual arousal [203].
arousal, including indirect heat dissipation mea- However, all these objective measures are rarely
sures (vaginal thermistors, labial thermistors, performed in clinical practice and lack of valida-
heated oxygen electrode), clitoral and vaginal tion, normative values, and correlation with sub-
photoplethysmography, Doppler ultrasonogra- jective response [205].
phy, dynamic contrast and non-contrast MRI, and In summary, the preliminary assessment of a
pudendal arteriogram [205]. Vaginal photople- woman with diabetes and FSD is based on vali-
thysmography is low cost and easy to use [205]. dated self-questionnaires, careful medical h­ istory
Doppler ultrasonography is a quick, relatively and physical examination, and routine laboratory
easy, non-invasive technique that provides a real-­ testing and hormonal status. Instrumental proce-
time assessment of anatomy and blood flow of dures, including several non-­validated methods,
genitalia [205]. Women with type 1 diabetes remain very limited in clinical practice (Fig. 6).

Diagnosis Medications list and psychological assessment


Medical and sexual history
1st Step Validated self-questionnaires (e.g., FSFI-19; FSFI-6)

General habitus and genitalia, anthropometric measures and


Physical examination BP, search for infection, inflammation, neoplasm, vaginal
atrophy, lesions

2nd Step Laboratory tests Metabolic, liver and kidney markers, hormonal status

Non-validated
methods Vaginal photoplethysmography, Doppler ultrasonography,
3rd Step Instrumental procedures
Selected techniques for neuromuscular system and genital blood flow
patients

Treatment
General Measures Lifestyle interventions Physical activity, diet, weight loss

Glucometabolic control Optimization of metabolic indices and comorbidities

Specific Measures Psychosexual strategies Psychological counseling, partner involvement

Post-menopausal
women Hormone therapy

Pharmacological strategies
Non-FDA PDE5I
approved Flibanserin
Ospemifene

Fig. 6  An algorithm for a diagnostic and therapeutic and comorbidities. Psychological counseling is a useful
approach to female sexual dysfunction. A preliminary tool for an underlying psychosexual dysfunction and
assessment is based on validated self-questionnaires, should include the partner. Hormone therapy is approved
medical history, and physical examination. A second step for postmenopausal women, whereas other pharmacologi-
concerns routine laboratory testing, while instrumental cal options are not FDA approved [193, 207]. FSFI female
procedures have limited application in clinical practice sexual function index, BP blood pressure, PDE5I type 5
[193, 205]. Therapeutic management is based on lifestyle phosphodiesterase inhibitors
changes and the optimization of glucometabolic control
520 V. Spallone et al.

5.5 Treatment of Diabetic Female the available studies have precluded the approval
Sexual Dysfunction of testosterone therapy for FSD. PDE5i-induced
vasodilatation might theoretically improve vagi-
Treatment of FSD in women with diabetes nal lubrication and vulvar engorgement, but
requires a multidisciplinary approach involving available results are limited and discordant: stud-
an endocrinologist, gynecologist, and psycho- ies using self-reported measures of sexual func-
therapeutic specialist. Psychological issues tioning showed mixed results, whereas major
should be identified and treated [193]. effects were reported by studies examining phys-
iological effects on genital vasocongestion [212].
5.5.1 Lifestyle Intervention The limited relevance of PDE5 vascular mecha-
Lifestyle changes, including a balanced diet, reg- nisms in FSD and low PDE5 levels in the female
ular exercise, and weight loss, should be the first reproductive system [194] are possible reasons
step for treating FSD in women with diabetes. for this. Other pharmacological options are sup-
The open MEDITA trial showed a lower decrease ported by promising data but not FDA approved.
in the FSFI score with a Mediterranean diet com- Flibanserin, a 5-HT1A agonist/5-HT2A antago-
pared to low-fat diet in women with newly diag- nist, showed an improvement in sexual desire and
nosed type 2 diabetes followed for 8 years [124]. reduction in distress associated with sexual dys-
Similarly, in the Look AHEAD trial, 1-year function in premenopausal women with hypoac-
intensive lifestyle intervention in women with tive sexual desire disorder [193]. Ospemifene, a
type 2 diabetes and FSD at baseline was associ- selective estrogen receptor modulator, was effec-
ated with greater weight loss and higher odds to tive in the treatment of vulvovaginal atrophy in
experience FSD remission than control manage- postmenopausal women with vaginal dryness
ment [208]. [193]. Additional strategies include pelvic phys-
iotherapy and counseling for sexual pain, and
5.5.2 Antihyperglycemic Treatment mindfulness and sex therapy for orgasmic dys-
Glycemic control by reducing the risk of diabetes-­ function [207].
related complications (including genitourinary In summary, therapeutic management is based
infections) is crucial [148]. Metformin is known on general measures including lifestyle changes
to provide a beneficial effect on all female sexual and optimization of glucometabolic control and
function domains, with this effect’s strength comorbidities. Psychological counseling is a use-
depending on the degree of insulin resistance ful tool for an underlying psychosexual dysfunc-
[209]. Given the positive impact of novel GLP-­ tion and should include the partner. Hormone
1RA and SGLT2i on body weight, it might be therapy is approved for postmenopausal women,
expected that these drugs would result in better whereas other pharmacological options are not
sexual outcomes, but no studies have evaluated FDA approved. A multidisciplinary approach to
this point in women and the increased risk of uro- FSD favors better communication and outcome
genital infections with SGLT2i should be taken (Fig. 6).
into account [176].

5.5.3 Pharmacological Options 6 Conclusions on Diabetic


Hormone therapy is approved for postmeno- Genitourinary Dysfunction
pausal women mainly in the form of vaginal
estrogen [193]. Although testosterone showed a Bladder and sexual dysfunction in diabetes are
beneficial effect on sexual function [210] and sci- present in more than 50% of patients with diabe-
entific societies suggest its evidence-based use tes. They represent a significant burden on
for postmenopausal women with hypoactive sex- patients’ mental health, QoL, and prognosis.
ual desire disorder with the exclusion of women Their multifactorial pathophysiology and multi-
at high cardiometabolic risk [211], limitations in dimensional characteristics call for more research
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 521

and an integrated multidisciplinary approach. EDIC Study Group. Burden of urological com-
plications in men and women with long-standing
They have been on the fringes of diabetes type 1 diabetes in the diabetes control and com-
research and clinical practice even more than dia- plications trial/epidemiology of diabetes interven-
betic neuropathy. Their clinical relevance, the tions and complications cohort. Diabetes Care.
ease of screening, and the availability of effective 2018;41(10):2170–7.
11. Michel MC, Mehlburger L, Schumacher H, Bressel
therapeutic options no longer justify them being HU, Goepel M. Effect of diabetes on lower urinary
left on the margins of the diabetic field. tract symptoms in patients with benign prostatic
hyperplasia. J Urol. 2000;163(6):1725–9.
12. Lee WC, Wu HP, Tai TY, Liu SP, Chen J, Yu
HJ. Effects of diabetes on female voiding behavior. J
References Urol. 2004;172(3):989–92.
13. Yu HJ, Lee WC, Liu SP, Tai TY, Wu HP, Chen
1. Frimodt-Møller C.  Diabetic cystopathy: epide- J.  Unrecognized voiding difficulty in female
miology and related disorders. Ann Intern Med. type 2 diabetic patients in the diabetes clinic: a
1980;92(2 Pt 2):318–21. prospective case-control study. Diabetes Care.
2. Brown JS, Wessells H, Chancellor MB, Howards 2004;27(4):988–9.
SS, Stamm WE, Stapleton AE, Steers WD, Van Den 14. Jackson RA, Vittinghoff E, Kanaya AM, Miles
Eeden SK, McVary KT.  Urologic complications of TP, Resnick HE, Kritchevsky SB, Simonsick EM,
diabetes. Diabetes Care. 2005;28(1):177–85. Brown JS, Health, Aging, and Body Composition
3. Kaplan SA, Te AE, Blaivas JG.  Urodynamic find- Study. Urinary incontinence in elderly women: find-
ings in patients with diabetic cystopathy. J Urol. ings from the Health, Aging, and Body Composition
1995;153(2):342–4. Study. Obstet Gynecol. 2004;104(2):301–7.
4. Thüroff JW, Abrams P, Andersson KE, Artibani 15. Lifford KL, Curhan GC, Hu FB, Barbieri RL,
W, Chapple CR, Drake MJ, Hampel C, Neisius A, Grodstein F.  Type 2 diabetes mellitus and risk of
Schröder A, Tubaro A.  EAU guidelines on urinary developing urinary incontinence. J Am Geriatr Soc.
incontinence. Eur Urol. 2011;59(3):387–400. 2005;53(11):1851–7.
5. Martin S, Vincent A, Taylor AW, Atlantis E, 16. Danforth KN, Townsend MK, Curhan GC, Resnick
Jenkins A, Januszewski A, O’Loughlin P, Wittert NM, Grodstein F. Type 2 diabetes mellitus and risk
G.  Lower urinary tract symptoms, depression, of stress, urge and mixed urinary incontinence. J
anxiety and systemic inflammatory factors in Urol. 2009;181(1):193–7.
men: a population-based cohort study. PLoS One. 17. Ebbesen MH, Hannestad YS, Midthjell K, Hunskaar
2015;10(10):e0137903. S.  Diabetes and urinary incontinence  – prevalence
6. Gacci M, Corona G, Sebastianelli A, Serni S, De data from Norway. Acta Obstet Gynecol Scand.
Nunzio C, Maggi M, Vignozzi L, Novara G, McVary 2007;86(10):1256–62.
KT, Kaplan SA, Gravas S, Chapple C.  Male lower 18. Phelan S, Kanaya AM, Subak LL, Hogan PE,
urinary tract symptoms and cardiovascular events: Espeland MA, Wing RR, Burgio KL, Dilillo V,
a systematic review and meta-analysis. Eur Urol. Gorin AA, West DS, Brown JS.  Action for Health
2016;70(5):788–96. in Diabetes (Look AHEAD) Research Group.
7. Joseph MA, Harlow SD, Wei JT, Sarma AV, Dunn Prevalence and risk factors for urinary incontinence
RL, Taylor JM, James SA, Cooney KA, Doerr KM, in overweight and obese diabetic women: action for
Montie JE, Schottenfeld D.  Risk factors for lower health in diabetes (Look AHEAD) study. Diabetes
urinary tract symptoms in a population-based sam- Care. 2009;32(8):1391–7.
ple of African-American men. Am J Epidemiol. 19. Appa AA, Brown JS, Creasman J, Van Den Eeden
2003;157(10):906–14. SK, Subak LL, Thom DH, Ferrara A, Huang
8. Liu RT, Chung MS, Lee WC, Chang SW, Huang ST, AJ. Clinical predictors and significance of postvoid
Yang KD, Chancellor MB, Chuang YC. Prevalence residual volume in women with diabetes. Diabetes
of overactive bladder and associated risk factors Res Clin Pract. 2013;101(2):164–9.
in 1359 patients with type 2 diabetes. Urology. 20. Hotaling JM, Sarma AV, Patel DP, Braffett BH, Cleary
2011;78(5):1040–5. PA, Feldman E, Herman WH, Martin CL, Jacobson
9. Pop-Busui R, Hotaling J, Braffett BH, Cleary PA, AM, Wessells H, Pop-Busui R.  Diabetes Control
Dunn RL, Martin CL, Jacobson AM, Wessells and Complications Trial/Epidemiology of Diabetes
H, Sarma AV.  DCCT/EDIC Research Group. Interventions and Complications Research Group.
Cardiovascular autonomic neuropathy, erectile dys- Cardiovascular autonomic neuropathy, sexual dys-
function and lower urinary tract symptoms in men function, and urinary incontinence in women with
with type 1 diabetes: findings from the DCCT/ type 1 diabetes. Diabetes Care. 2016;39(9):1587–93.
EDIC. J Urol. 2015;193(6):2045–51. 21. Lenherr SM, Clemens JQ, Braffett BH, Dunn
10. Wessells H, Braffett BH, Holt SK, Jacobson AM, RL, Cleary PA, Kim C, Herman WH, Hotaling
Kusek JW, Cowie C, Dunn RL, Sarma AV. DCCT/ JM, Jacobson AM, Brown JS, Wessells H, Sarma
522 V. Spallone et al.

AV.  DCCT/EDIC Research Group. Glycaemic diabetes-associated bladder complications and novel
control and risk of incident urinary incontinence therapy. BJU Int. 2005;95(6):733–8.
in women with Type 1 diabetes: results from the 34. Mossa AH, Galan A, Cammisotto PG, Velasquez
Diabetes Control and Complications Trial and Flores M, Shamout S, Barcelona P, Saragovi HU,
Epidemiology of Diabetes Interventions and Campeau L. Antagonism of proNGF or its receptor
Complications (DCCT/EDIC) study. Diabet Med. p75NTR reverses remodelling and improves bladder
2016;33(11):1528–35. function in a mouse model of diabetic voiding dys-
22. Wilke T, Boettger B, Berg B, Groth A, Mueller function. Diabetologia. 2020;63(9):1932–46.
S, Botteman M, Yu S, Fuchs A, Maywald 35. Daneshgari F, Liu G, Hanna-Mitchell AT.  Path of
U. Epidemiology of urinary tract infections in type translational discovery of urological complications
2 diabetes mellitus patients: an analysis based on a of obesity and diabetes. Am J Physiol Ren Physiol.
large sample of 456,586 German T2DM patients. J 2017;312(5):F887–96.
Diabetes Complicat. 2015;29(8):1015–23. 36. de Oliveira MG, de Medeiros ML, Tavares EBG,
23. Renko M, Tapanainen P, Tossavainen P, Pokka Mónica FZ, Antunes E.  Methylglyoxal, a reactive
T, Uhari M.  Meta-analysis of the significance of glucose metabolite, induces bladder overactivity
asymptomatic bacteriuria in diabetes. Diabetes Care. in addition to inflammation in mice. Front Physiol.
2011;34(1):230–5. 2020;11:290.
24. Boyko EJ, Fihn SD, Scholes D, Chen CL, Normand 37. Klee NS, McCarthy CG, Lewis S, McKenzie JL,
EH, Yarbro P.  Diabetes and the risk of acute uri- Vincent JE, Webb RC. Urothelial senescence in the
nary tract infection among postmenopausal women. pathophysiology of diabetic bladder dysfunction-a
Diabetes Care. 2002;25(10):1778–83. novel hypothesis. Front Surg. 2018;5:72.
25. de Groat WC, Yoshimura N. Anatomy and physiol- 38. Geerlings SE, Meiland R, van Lith EC, Brouwer
ogy of the lower urinary tract. Handb Clin Neurol. EC, Gaastra W, Hoepelman AI.  Adherence of type
2015;130:61–108. 1-­fimbriated Escherichia coli to uroepithelial cells:
26. Daneshgari F, Liu G, Birder L, Hanna-Mitchell more in diabetic women than in control subjects.
AT, Chacko S.  Diabetic bladder dysfunction: Diabetes Care. 2002;25(8):1405–9.
­current translational knowledge. J Urol. 2009;182(6 39. Yang Z, Dolber PC, Fraser MO. Diabetic urethropa-
Suppl):S18–26. thy compounds the effects of diabetic cystopathy. J
27. Esteghamati A, Rashidi A, Nikfallah A, Urol. 2007;178(5):2213–9.
Yousefizadeh A.  The association between urody- 40. Cao N, Gu B, Gotoh D, Yoshimura N.  Time-­
namic findings and microvascular complications in dependent changes of urethral function in diabetes
patients with long-term type 2 diabetes but with- mellitus: a review. Int Neurourol J. 2019;23(2):91–9.
out voiding symptoms. Diabetes Res Clin Pract. 41. Beshay E, Carrier S. Oxidative stress plays a role in
2007;78(1):42–50. diabetes-induced bladder dysfunction in a rat model.
28. Rapidi CA, Karandreas N, Katsifotis C, Benroubi Urology. 2004;64(5):1062–7.
M, Petropoulou K, Theodorou C. A combined uro- 42. Yuan Z, Tang Z, He C, Tang W. Diabetic cystopathy:
dynamic and electrophysiological study of diabetic a review. J Diabetes. 2015;7(4):442–7.
cystopathy. Neurourol Urodyn. 2006;25(1):32–8. 43. Harding GK, Zhanel GG, Nicolle LE, Cheang
29. Kempler P, Amarenco G, Freeman R, Frontoni M. Manitoba Diabetes Urinary Tract Infection Study
S, Horowitz M, Stevens M, Low P, Pop-Busui R, Group. Antimicrobial treatment in diabetic women
Tahrani AA, Tesfaye S, Várkonyi T, Ziegler D, with asymptomatic bacteriuria. N Engl J Med.
Valensi P.  Toronto Consensus Panel on Diabetic 2002;347(20):1576–83.
Neuropathy. Management strategies for gastrointes- 44. Nicolle LE, Gupta K, Bradley SF, Colgan R, DeMuri
tinal, erectile, bladder, and sudomotor dysfunction GP, Drekonja D, Eckert LO, Geerlings SE, Köves B,
in patients with diabetes. Diabetes Metab Res Rev. Hooton TM, Juthani-Mehta M, Knight SL, Saint
2011;27(7):665–77. S, Schaeffer AJ, Trautner B, Wullt B, Siemieniuk
30. Van Poppel H, Stessens R, Van Damme B, Carton R.  Clinical practice guideline for the management
H, Baert L. Diabetic cystopathy: neuropathological of asymptomatic bacteriuria: 2019 update by the
examination of urinary bladder biopsies. Eur Urol. Infectious Diseases Society of America. Clin Infect
1988;15(1–2):128–31. Dis. 2019;68(10):1611–5.
31. Lee WC, Wu HP, Tai TY, Yu HJ, Chiang 45. Tang H, Cui W, Li D, Wang T, Zhang J, Zhai S,
PH.  Investigation of urodynamic characteristics Song Y.  Sodium-glucose co-transporter 2 inhibi-
and bladder sensory function in the early stages of tors in addition to insulin therapy for management
diabetic bladder dysfunction in women with type 2 of type 2 diabetes mellitus: a meta-analysis of ran-
diabetes. J Urol. 2009;181(1):198–203. domized controlled trials. Diabetes Obes Metab.
32. de Groat WC, Griffiths D, Yoshimura N.  Neural 2017;19(1):142–7.
control of the lower urinary tract. Compr Physiol. 46. Donnan JR, Grandy CA, Chibrikov E, Pharm DCM,
2015;5(1):327–96. Aubrey-Bassler K, Johnston K, Swab M, Hache J,
33. Yoshimura N, Chancellor MB, Andersson KE, Christ Curnew D, Nguyen H, Gamble JM. Dose response
GJ. Recent advances in understanding the biology of of sodium glucose cotransporter-2 inhibitors in rela-
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 523

tion to urinary tract infections: a systematic review 58. Abrams P, Avery K, Gardener N, Donovan J. ICIQ
and network meta-analysis of randomized controlled Advisory Board. The international consultation
trials. CMAJ Open. 2018;6(4):E594–602. on incontinence modular questionnaire. J Urol.
47. U.S.  Food and Drug Administration. Drug safety 2006;175(3 Pt 1):1063–6; www.iciq.net.
communication: FDA revises labels of SGLT2 59. Greco C, Di Gennaro F, D’Amato C, Morganti R,
inhibitors for diabetes to include warnings about Corradini D, Sun A, Longo S, Lauro D, Pierangeli G,
too much acid in the blood and serious uri- Cortelli P, Spallone V. Validation of the Composite
nary tract infections. https://www.fda.gov/drugs/ Autonomic Symptom Score 31 (COMPASS 31)
drug-­s afety-­a nd-­availability/fda-­r evises-­l abels-­ for the assessment of symptoms of autonomic
sglt2-­inhibitors-­diabetes-­include-­warnings-­about-­ neuropathy in people with diabetes. Diabet Med.
too-­much-­acid-­blood-­and-­serious. Accessed 23 Feb 2017;34(6):834–8.
2021. 60. Pop-Busui R, Boulton AJ, Feldman EL, Bril
48. Yap TL, Cromwell DC, Emberton M. A systematic V, Freeman R, Malik RA, Sosenko JM, Ziegler
review of the reliability of frequency-volume charts D.  Diabetic neuropathy: a position statement by
in urological research and its implications for the the American Diabetes Association. Diabetes Care.
optimum chart duration. BJU Int. 2007;99(1):9–16. 2017;40(1):136–54.
49. van Haarst EP, Bosch JL.  The optimal duration of 61. Phelan S, Kanaya AM, Ma Y, Vittinghoff E, Barrett-­
frequency-volume charts related to compliance and Connor E, Wing R, Kusek JW, Orchard TJ, Crandall
reliability. Neurourol Urodyn. 2014;33(3):296–301. JP, Montez MG, Brown JS.  Diabetes Prevention
50. Panicker JN, Fowler CJ, Kessler TM. Lower urinary Program Research Group. Long-term prevalence and
tract dysfunction in the neurological patient: clini- predictors of urinary incontinence among women in
cal assessment and management. Lancet Neurol. the Diabetes Prevention Program Outcomes Study.
2015;14(7):720–32. Int J Urol. 2015;22(2):206–12.
51. Rosier PFWM, Schaefer W, Lose G, Goldman 62. Breyer BN, Phelan S, Hogan PE, Rosen RC, Kitabchi
HB, Guralnick M, Eustice S, Dickinson T, AE, Wing RR, Brown JS.  Look AHEAD Research
Hashim H.  International Continence Society Good Group. Intensive lifestyle intervention reduces uri-
Urodynamic Practices and Terms 2016: urodynam- nary incontinence in overweight/obese men with
ics, uroflowmetry, cystometry, and pressure-flow type 2 diabetes: results from the Look AHEAD trial.
study. Neurourol Urodyn. 2017;36(5):1243–60. J Urol. 2014;192(1):144–9.
52. Groen J, Pannek J, Castro Diaz D, Del Popolo 63. Corona G, Tirabassi G, Santi D, Maseroli E, Gacci
G, Gross T, Hamid R, Karsenty G, Kessler TM, M, Dicuio M, Sforza A, Mannucci E, Maggi
Schneider M, ‘t Hoen L, Blok B.  Summary of M. Sexual dysfunction in subjects treated with inhib-
European Association of Urology (EAU) guidelines itors of 5α-reductase for benign prostatic hyperpla-
on neuro-urology. Eur Urol. 2016;69(2):324–33. sia: a comprehensive review and meta-analysis.
53. Stöhrer M, Blok B, Castro-Diaz D, Chartier-Kastler Andrology. 2017;5(4):671–8.
E, Del Popolo G, Kramer G, Pannek J, Radziszewski 64. Han HH, Ko WJ, Yoo TK, Oh TH, Kim DY, Kwon
P, Wyndaele JJ. EAU guidelines on neurogenic lower DD, Byun SS, Kim SI, Jung TY. Factors associated
urinary tract dysfunction. Eur Urol. 2009;56(1):81–8. with continuing medical therapy after transurethral
54. Dumoulin C, Adewuyi T, Booth J, Bradley C, Burgio resection of prostate. Urology. 2014;84(3):675–80.
K, Hagen S, Hunter K, Imamura M, Morin M, 65. Schneider T, Marschall-Kehrel D, Hanisch JU,
Morkved S, Thakar R, Wallace S, Williams K. Adult Michel MC. Does concomitant diabetes affect treat-
conservative management. In: Abrams P, Cardozo ment responses in overactive bladder patients? Int J
L, Wagg A, Wein A, editors. Incontinence: 6th Clin Pract. 2013;67(11):1138–43.
International consultation on incontinence, Tokyo, 66. Wagg A, Nitti VW, Kelleher C, Castro-Diaz D,
September 2016. Bristol: International Continence Siddiqui E, Berner T.  Oral pharmacotherapy for
Society (ICS) and International Consultation on overactive bladder in older patients: mirabegron as
Urological Diseases (ICUD); 2017. p. 1443–628. a potential alternative to antimuscarinics. Curr Med
55. Coolen RL, Groen J, Scheepe JR, Blok Res Opin. 2016;32(4):621–38.
BFM.  Transcutaneous electrical nerve stimulation 67. Finazzi-Agrò E, Petta F, Sciobica F, Pasqualetti
and percutaneous tibial nerve stimulation to treat P, Musco S, Bove P.  Percutaneous tibial nerve
idiopathic nonobstructive urinary retention: a sys- stimulation effects on detrusor overactivity incon-
tematic review. Eur Urol Focus. 2020;7(5):1184–94. tinence are not due to a placebo effect: a random-
56. Wittig L, Carlson KV, Andrews JM, Crump RT, ized, double-blind, placebo controlled trial. J Urol.
Baverstock RJ.  Diabetic bladder dysfunction: a 2010;184(5):2001–6.
review. Urology. 2019;123:1–6. 68. Peters KM, Macdiarmid SA, Wooldridge LS,
57. Coyne KS, Thompson CL, Lai JS, Sexton CC.  An Leong FC, Shobeiri SA, Rovner ES, Siegel SW,
overactive bladder symptom and health-related Tate SB, Jarnagin BK, Rosenblatt PL, Feagins
quality of life short-form: validation of the OAB-q BA.  Randomized trial of percutaneous tibial nerve
SF. Neurourol Urodyn. 2015;34(3):255–63. stimulation versus extended-release tolterodine:
524 V. Spallone et al.

results from the overactive bladder innovative ther- meta-analysis of 145 studies. Diabet Med.
apy trial. J Urol. 2009;182(3):1055–61. 2017;34(9):1185–92.
69. Gaziev G, Topazio L, Iacovelli V, Asimakopoulos 81. Fedele D, Coscelli C, Santeusanio F, Bortolotti
A, Di Santo A, De Nunzio C, Finazzi-Agrò A, Chatenoud L, Colli E, Landoni M, Parazzini
E.  Percutaneous Tibial Nerve Stimulation (PTNS) F. Erectile dysfunction in diabetic subjects in Italy.
efficacy in the treatment of lower urinary tract Gruppo Italiano Studio Deficit Erettile nei Diabetici.
dysfunctions: a systematic review. BMC Urol. Diabetes Care. 1998;21(11):1973–7.
2013;13:61. 82. Parazzini F, Menchini Fabris F, Bortolotti A, Calabrò
70. Daniels DH, Powell CR, Braasch MR, Kreder A, Chatenoud L, Colli E, Landoni M, Lavezzari M,
KJ.  Sacral neuromodulation in diabetic patients: Turchi P, Sessa A, Mirone V. Frequency and deter-
success and complications in the treatment minants of erectile dysfunction in Italy. Eur Urol.
of voiding dysfunction. Neurourol Urodyn. 2000;37(1):43–9.
2010;29(4):578–81. 83. Corona G, Giorda CB, Cucinotta D, Guida P, Nada
71. Haraway AM, Clemens JQ, He C, Stroup C, Atiemo E. Gruppo di studio SUBITO-DE. Sexual dysfunc-
HO, Cameron AP.  Differences in sacral neuro- tion at the onset of type 2 diabetes: the interplay of
modulation device infection rates based on pre- depression, hormonal and cardiovascular factors. J
operative antibiotic selection. Int Urogynecol J. Sex Med. 2014;11(8):2065–73.
2013;24(12):2081–5. 84. Skeldon SC, Detsky AS, Goldenberg SL, Law
72. Wang CC, Liao CH, Kuo HC.  Diabetes mellitus MR. Erectile dysfunction and undiagnosed diabetes,
does not affect the efficacy and safety of intravesi- hypertension, and hypercholesterolemia. Ann Fam
cal onabotulinumtoxinA injection in patients with Med. 2015;13(4):331–5.
refractory detrusor overactivity. Neurourol Urodyn. 85. Johannes CB, Araujo AB, Feldman HA, Derby CA,
2014;33(8):1235–9. Kleinman KP, McKinlay JB.  Incidence of erectile
73. Lamin E, Newman DK.  Clean intermittent dysfunction in men 40 to 69 years old: longitudinal
catheterization revisited. Int Urol Nephrol. results from the Massachusetts male aging study. J
2016;48(6):931–9. Urol. 2000;163(2):460–3.
74. Pistolesi D, Barsotti M, Giannarini G, Mantovani 86. Fedele D, Coscelli C, Cucinotta D, Forti G,
M, Boggi U, Kessler TM, Selli C.  Sacral neuro- Santeusanio F, Viaggi S, Fiori G, Velonà T,
modulation enabling simultaneous pancreas and Lavezzari M. Diade Study Group. Incidence of erec-
kidney transplantation: a first case. BMJ Case Rep. tile dysfunction in Italian men with diabetes. J Urol.
2009;2009:bcr08.2008.0823. 2001;166(4):1368–71.
75. Bump RC, Voss S, Beardsworth A, Manning M, 87. Braun M, Wassmer G, Klotz T, Reifenrath B,
Zhao YD, Chen W. Long-term efficacy of duloxetine Mathers M, Engelmann U. Epidemiology of erectile
in women with stress urinary incontinence. BJU Int. dysfunction: results of the ‘Cologne Male Survey’.
2008;102(2):214–8. Int J Impot Res. 2000;12(6):305–11.
76. Kokanali MK, Doğanay M, Aksakal O, Cavkaytar 88. Nicolosi A, Moreira ED Jr, Shirai M, Bin Mohd
S, Topçu HO, Özer İ. Risk factors for mesh ero- Tambi MI, Glasser DB.  Epidemiology of erectile
sion after vaginal sling procedures for urinary dysfunction in four countries: cross-national study
incontinence. Eur J Obstet Gynecol Reprod Biol. of the prevalence and correlates of erectile dysfunc-
2014;177:146–50. tion. Urology. 2003;61(1):201–6.
77. Corona G, Isidori AM, Aversa A, Bonomi M, Ferlin 89. Malavige LS, Levy JC. Erectile dysfunction in dia-
A, Foresta C, La Vignera S, Maggi M, Pivonello R, betes mellitus. J Sex Med. 2009;6(5):1232–47.
Vignozzi L, Lombardo F.  Male and female sexual 90. Sarma AV, Hotaling JM, de Boer IH, Dunn RL,
dysfunction in diabetic subjects: focus on new anti- Oerline MK, Singh K, Goldberg J, Jacobson A,
hyperglycemic drugs. Rev Endocr Metab Disord. Braffett B, Herman WH, Pop-Busui R, Wessells
2020;21(1):57–65. H.  DCCT/EDIC Research Group. Blood pressure,
78. McCabe MP, Sharlip ID, Atalla E, Balon R, Fisher antihypertensive medication use, and risk of erectile
AD, Laumann E, Lee SW, Lewis R, Segraves dysfunction in men with type I diabetes. J Hypertens.
RT.  Definitions of sexual dysfunctions in women 2019;37(5):1070–6.
and men: a consensus statement from the Fourth 91. Rosen RC, Wing RR, Schneider S, Wadden TA,
International Consultation on Sexual Medicine Foster GD, West DS, Kitabchi AE, Brancati FL,
2015. J Sex Med. 2016;13(2):135–43. Maschak-Carey BJ, Bahnson JL, Lewis CE,
79. Feldman HA, Goldstein I, Hatzichristou DG, Krane Gendrano Iii IN. Erectile dysfunction in type 2 dia-
RJ, McKinlay JB.  Impotence and its medical and betic men: relationship to exercise fitness and car-
psychosocial correlates: results of the Massachusetts diovascular risk factors in the Look AHEAD trial. J
Male Aging Study. J Urol. 1994;151:54–61. Sex Med. 2009;6(5):1414–22.
80. Kouidrat Y, Pizzol D, Cosco T, Thompson T, 92. Liu Q, Zhang Y, Wang J, Li S, Cheng Y, Guo J, Tang
Carnaghi M, Bertoldo A, Solmi M, Stubbs B, Y, Zeng H, Zhu Z. Erectile dysfunction and depres-
Veronese N.  High prevalence of erectile dys- sion: a systematic review and meta-analysis. J Sex
function in diabetes: a systematic review and Med. 2018;15(8):1073–82.
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 525

93. Pouwer F. Depression: a common and burdensome 104. Montorsi P, Ravagnani PM, Galli S, Rotatori F,
complication of diabetes that warrants the continued Briganti A, Salonia A, Rigatti P, Montorsi F.  The
attention of clinicians, researchers and healthcare artery size hypothesis: a macrovascular link between
policy makers. Diabetologia. 2017;60(1):30–4. erectile dysfunction and coronary artery disease. Am
94. De Berardis G, Franciosi M, Belfiglio M, Di Nardo J Cardiol. 2005;96(12B):19M–23M.
B, Greenfield S, Kaplan SH, Pellegrini F, Sacco 105. Frantzen J, Speel TG, Kiemeney LA, Meuleman
M, Tognoni G, Valentini M, Nicolucci A.  Quality EJ.  Cardiovascular risk among men seeking
of Care and Outcomes in Type 2 Diabetes (QuED) help for erectile dysfunction. Ann Epidemiol.
Study Group. Erectile dysfunction and qual- 2006;16(2):85–90.
ity of life in type 2 diabetic patients: a serious 106. Gazzaruso C, Solerte SB, Pujia A, Coppola A,
problem too often overlooked. Diabetes Care. Vezzoli M, Salvucci F, Valenti C, Giustina A,
2002;25(2):284–91. Garzaniti A.  Erectile dysfunction as a predictor of
95. De Berardis G, Pellegrini F, Franciosi M, Belfiglio cardiovascular events and death in diabetic patients
M, Di Nardo B, Greenfield S, Kaplan SH, Rossi with angiographically proven asymptomatic coro-
MC, Sacco M, Tognoni G, Valentini M, Nicolucci nary artery disease: a potential protective role for
A.  QuED (Quality of Care and Outcomes in Type statins and 5-phosphodiesterase inhibitors. J Am
2 Diabetes) Study Group. Longitudinal assessment Coll Cardiol. 2008;51(21):2040–4.
of quality of life in patients with type 2 diabetes and 107. Santi D, Granata AR, Guidi A, Pignatti E, Trenti
self-reported erectile dysfunction. Diabetes Care. T, Roli L, Bozic R, Zaza S, Pacchioni C, Romano
2005;28(11):2637–43. S, Nofer JR, Rochira V, Carani C, Simoni M.  Six
96. Jacobson AM, Braffett BH, Cleary PA, Dunn months of daily treatment with vardenafil improves
RL, Larkin ME, Wessells H, Sarma AV.  DCCT/ parameters of endothelial inflammation and of
EDIC Research Group. Relationship of urologic hypogonadism in male patients with type 2 dia-
complications with health-related quality of life betes and erectile dysfunction: a randomized,
and perceived value of health in men and women double-blind, prospective trial. Eur J Endocrinol.
with type 1 diabetes: the Diabetes Control and 2016;174(4):513–22.
Complications Trial/Epidemiology of Interventions 108. Tzoumas N, Farrah TE, Dhaun N, Webb
and Complications (DCCT/EDIC) cohort. Diabetes DJ.  Established and emerging therapeutic uses of
Care. 2015;38(10):1904–12. PDE type 5 inhibitors in cardiovascular disease. Br J
97. Corona G, Rastrelli G, Isidori AM, Pivonello Pharmacol. 2020;177(24):5467–88.
R, Bettocchi C, Reisman Y, Sforza A, Maggi 109. Pavy-Le Traon A, Fontaine S, Tap G, Guidolin B,
M.  Erectile dysfunction and cardiovascular risk: a Senard JM, Hanaire H.  Cardiovascular autonomic
review of current findings. Expert Rev Cardiovasc neuropathy and other complications in type 1 diabe-
Ther. 2020;18(3):155–64. tes. Clin Auton Res. 2010;20(3):153–60.
98. Besiroglu H, Otunctemur A, Ozbek E. The relation- 110. Wessells H, Penson DF, Cleary P, Rutledge BN,
ship between metabolic syndrome, its components, Lachin JM, McVary KT, Schade DS, Sarma
and erectile dysfunction: a systematic review and a AV.  Diabetes Control and Complications Trial/
meta-analysis of observational studies. J Sex Med. Epidemiology of Diabetes Interventions and
2015;12(6):1309–18. Complications Research Group. Effect of intensive
99. Ning L, Yang L.  Hypertension might be a risk glycemic therapy on erectile function in men with
factor for erectile dysfunction: a meta-analysis. type 1 diabetes. J Urol. 2011;185(5):1828–34.
Andrologia. 2017;49(4) 111. Azmi S, Ferdousi M, Alam U, Petropoulos IN,
100. Wang XM, Bai YJ, Yang YB, Li JH, Tang Y, Han Ponirakis G, Marshall A, Asghar O, Fadavi H, Jones
P. Alcohol intake and risk of erectile dysfunction: a W, Tavakoli M, Boulton AJM, Jeziorska M, Soran H,
dose-response meta-analysis of observational stud- Efron N, Malik RA. Small-fibre neuropathy in men
ies. Int J Impot Res. 2018;30(6):342–51. with type 1 diabetes and erectile dysfunction: a cross-­
101. Allen MS, Walter EE.  Erectile dysfunction: an sectional study. Diabetologia. 2017;60(6):1094–101.
umbrella review of meta-analyses of risk-factors, 112. Dhage S, Ho JH, Ferdousi M, Kalteniece A, Azmi S,
treatment, and prevalence outcomes. J Sex Med. Adam S, Marshall A, Jeziorska M, Donn R, Soran H,
2019;16(4):531–41. Malik RA. Small fibre pathology is associated with
102. Zhao B, Hong Z, Wei Y, Yu D, Xu J, Zhang W. Erectile erectile dysfunction in men with type 2 diabetes.
dysfunction predicts cardiovascular events as an Diabetes Metab Res Rev. 2020;36(3):e3263.
independent risk factor: a systematic review and 113. Wellmer A, Sharief MK, Knowles CH, Misra VP,
meta-analysis. J Sex Med. 2019;16(7):1005–17. Kopelman P, Ralph D, Anand P. Quantitative sensory
103. Vlachopoulos CV, Terentes-Printzios DG, and autonomic testing in male diabetic patients with
Ioakeimidis NK, Aznaouridis KA, Stefanadis erectile dysfunction. BJU Int. 1999;83(1):66–70.
CI. Prediction of cardiovascular events and all-cause 114. Sáenz de Tejada I, Angulo J, Cellek S, González-­
mortality with erectile dysfunction: a systematic Cadavid N, Heaton J, Pickard R, Simonsen
review and meta-analysis of cohort studies. Circ U.  Pathophysiology of erectile dysfunction. J Sex
Cardiovasc Qual Outc. 2013;6(1):99–109. Med. 2005;2(1):26–39.
526 V. Spallone et al.

115. Saenz de Tejada I, Goldstein I, Azadzoi K, Krane RJ, KT, Miner MM, Nelson CJ, Sadeghi-Nejad H, Seftel
Cohen RA.  Impaired neurogenic and endothelium-­ AD, Shindel AW. Erectile dysfunction: AUA guide-
mediated relaxation of penile smooth muscle line. J Urol. 2018;200(3):633–41.
from diabetic men with impotence. N Engl J Med. 131. Nehra A, Jackson G, Miner M, Billups KL, Burnett
1989;320(16):1025–30. AL, Buvat J, Carson CC, Cunningham GR, Ganz P,
116. Ganz P.  Erectile dysfunction: pathophysiologic Goldstein I, Guay AT, Hackett G, Kloner RA, Kostis
mechanisms pointing to underlying cardiovascular J, Montorsi P, Ramsey M, Rosen R, Sadovsky R,
disease. Am J Cardiol. 2005;96(12B):8M–12M. Seftel AD, Shabsigh R, Vlachopoulos C, Wu FC. The
117. Andersson KE, Wagner G.  Physiology of penile Princeton III Consensus recommendations for the
erection. Physiol Rev. 1995;75(1):191–236. management of erectile dysfunction and cardiovas-
118. Dean RC, Lue TF. Physiology of penile erection and cular disease. Mayo Clin Proc. 2012;87(8):766–78.
pathophysiology of erectile dysfunction. Urol Clin 132. Althof SE, Rosen RC, Perelman MA, Rubio-­
North Am. 2005;32(4):379–95. Aurioles E.  Standard operating procedures for tak-
119. Isidori AM, Buvat J, Corona G, Goldstein I, Jannini ing a sexual history. J Sex Med. 2013;10(1):26–35.
EA, Lenzi A, Porst H, Salonia A, Traish AM, Maggi 133. Cappelleri JC, Rosen RC.  The Sexual Health
M. A critical analysis of the role of testosterone in Inventory for Men (SHIM): a 5-year review of
erectile function: from pathophysiology to treatment- research and clinical experience. Int J Impot Res.
­a systematic review. Eur Urol. 2014;65(1):99–112. 2005;17(4):307–19.
120. Sopko NA, Hannan JL, Bivalacqua TJ. Understanding 134. Petrone L, Mannucci E, Corona G, Bartolini M,
and targeting the Rho kinase pathway in erectile dys- Forti G, Giommi R, Maggi M. Structured interview
function. Nat Rev Urol. 2014;11(11):622–8. on erectile dysfunction (SIEDY): a new, multidi-
121. Ludwig W, Phillips M. Organic causes of erectile dys- mensional instrument for quantification of pathoge-
function in men under 40. Urol Int. 2014;92(1):1–6. netic issues on erectile dysfunction. Int J Impot Res.
122. Jannini EA, McCabe MP, Salonia A, Montorsi 2003;15(3):210–20.
F, Sachs BD.  Organic vs. psychogenic? The 135. Ghanem HM, Salonia A, Martin-Morales A.  SOP:
Manichean diagnosis in sexual medicine. J Sex Med. physical examination and laboratory testing
2010;7(5):1726–33. for men with erectile dysfunction. J Sex Med.
123. Gupta BP, Murad MH, Clifton MM, Prokop L, Nehra 2013;10(1):108–10.
A, Kopecky SL. The effect of lifestyle modification 136. Gratzke C, Bachmann A, Descazeaud A, Drake MJ,
and cardiovascular risk factor reduction on erectile Madersbacher S, Mamoulakis C, Oelke M, Tikkinen
dysfunction: a systematic review and meta-analysis. KAO, Gravas S. EAU guidelines on the assessment
Arch Intern Med. 2011;171(20):1797–803. of non-neurogenic male lower urinary tract symp-
124. Maiorino MI, Bellastella G, Giugliano D, Esposito toms including benign prostatic obstruction. Eur
K. From inflammation to sexual dysfunctions: a jour- Urol. 2015;67(6):1099–109.
ney through diabetes, obesity, and metabolic syn- 137. Spallone V, Ziegler D, Freeman R, Bernardi L,
drome. J Endocrinol Investig. 2018;41(11):1249–58. Frontoni S, Pop-Busui R, Stevens M, Kempler P,
125. Dhindsa S, Ghanim H, Batra M, Dandona Hilsted J, Tesfaye S, Low P, Valensi P.  Toronto
P.  Hypogonadotropic hypogonadism in men with Consensus Panel on Diabetic Neuropathy.
diabesity. Diabetes Care. 2018;41(7):1516–25. Cardiovascular autonomic neuropathy in diabetes:
126. Brüning JC, Gautam D, Burks DJ, Gillette J, clinical impact, assessment, diagnosis, and manage-
Schubert M, Orban PC, Klein R, Krone W, Müller-­ ment. Diabetes Metab Res Rev. 2011;27(7):639–53.
Wieland D, Kahn CR. Role of brain insulin receptor 138. Kamenov ZA.  A comprehensive review of erec-
in control of body weight and reproduction. Science. tile dysfunction in men with diabetes. Exp Clin
2000;289(5487):2122–5. Endocrinol Diabetes. 2015;123(3):141–58.
127. Isidori AM, Caprio M, Strollo F, Moretti C, Frajese 139. Corona G, Goulis DG, Huhtaniemi I, Zitzmann
G, Isidori A, Fabbri A.  Leptin and androgens in M, Toppari J, Forti G, Vanderschueren D, Wu
male obesity: evidence for leptin contribution to FC. European Academy of Andrology (EAA) guide-
reduced androgen levels. J Clin Endocrinol Metab. lines on investigation, treatment and monitoring
1999;84(10):3673–80. of functional hypogonadism in males: endorsing
128. Zhai L, Zhao J, Zhu Y, Liu Q, Niu W, Liu C, Wang organization: European Society of Endocrinology.
Y. Downregulation of leptin receptor and kisspeptin/ Andrology. 2020;8(5):970–87.
GPR54  in the murine hypothalamus contributes to 140. Bella AJ, Lee JC, Carrier S, Bénard F, Brock GB.
male hypogonadism caused by high-fat diet-induced 2015 CUA Practice guidelines for erectile dysfunc-
obesity. Endocrine. 2018;62(1):195–206. tion. Can Urol Assoc J. 2015;9(1-2):23–9.
129. Simó R, Sáez-López C, Barbosa-Desongles A, 141. Corona G, Fagioli G, Mannucci E, Romeo A,
Hernández C, Selva DM.  Novel insights in SHBG Rossi M, Lotti F, Sforza A, Morittu S, Chiarini V,
regulation and clinical implications. Trends Casella G, Di Pasquale G, Bandini E, Forti G, Maggi
Endocrinol Metab. 2015;26(7):376–83. M. Penile Doppler ultrasound in patients with erec-
130. Burnett AL, Nehra A, Breau RH, Culkin DJ, Faraday tile dysfunction (ED): role of peak systolic velocity
MM, Hakim LS, Heidelbaugh J, Khera M, McVary measured in the flaccid state in predicting arterio-
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 527

genic ED and silent coronary artery disease. J Sex 154. Liao X, Qiu S, Bao Y, Wang W, Yang L, Wei
Med. 2008;5(11):2623–34. Q.  Comparative efficacy and safety of phosphodi-
142. Jung DC, Park SY, Lee JY.  Penile Doppler esterase type 5 inhibitors for erectile dysfunction
ultrasonography revisited. Ultrasonography. in diabetic men: a Bayesian network meta-­analysis
2018;37(1):16–24. of randomized controlled trials. World J Urol.
143. Hatzimouratidis K, Salonia A, Adaikan G, Buvat J, 2019;37(6):1061–74.
Carrier S, El-Meliegy A, McCullough A, Torres LO, 155. Giannetta E, Feola T, Gianfrilli D, Pofi R, Dall’Armi
Khera M. Pharmacotherapy for erectile dysfunction: V, Badagliacca R, Barbagallo F, Lenzi A, Isidori
recommendations from the Fourth International AM. Is chronic inhibition of phosphodiesterase type
Consultation for Sexual Medicine (ICSM 2015). J 5 cardioprotective and safe? A meta-analysis of ran-
Sex Med. 2016;13(4):465–88. domized controlled trials. BMC Med. 2014;12:185.
144. Algeffari M, Jayasena CN, MacKeith P, Thapar 156. Liu B, Zhu L, Zhong J, Zeng G, Deng T. The asso-
A, Dhillo WS, Oliver N.  Testosterone therapy ciation between phosphodiesterase type 5 inhibitor
for sexual dysfunction in men with Type 2 dia- use and risk of non-arteritic anterior ischemic optic
betes: a systematic review and meta-analysis neuropathy: a systematic review and meta-analysis.
of randomized controlled trials. Diabet Med. Sex Med. 2018;6(3):185–92.
2018;35(2):195–202. 157. Manna S, Gray ML, Kaul VF, Wanna
145. Duncan C, Omran GJ, Teh J, Davis NF, Bolton DM, G.  Phosphodiesterase-5 (PDE-5) Inhibitors and
Lawrentschuk N.  Erectile dysfunction: a global ototoxicity: a systematic review. Otol Neurotol.
review of intracavernosal injectables. World J Urol. 2019;40(3):276–83.
2019;37(6):1007–14. 158. Giuliano F, Jackson G, Montorsi F, Martin-Morales
146. Levine LA, Becher EF, Bella AJ, Brant WO, A, Raillard P. Safety of sildenafil citrate: review of
Kohler TS, Martinez-Salamanca JI, Trost L, Morey 67 double-blind placebo-controlled trials and the
AF. Penile prosthesis surgery: current recommenda- postmarketing safety database. Int J Clin Pract.
tions from the international consultation on sexual 2010;64(2):240–55.
medicine. J Sex Med. 2016;13(4):489–518. 159. Lee M, Sharifi R.  Non-invasive management
147. Silva AB, Sousa N, Azevedo LF, Martins C. Physical options for erectile dysfunction when a phospho-
activity and exercise for erectile dysfunction: sys- diesterase type 5 inhibitor fails. Drugs Aging.
tematic review and meta-analysis. Br J Sports Med. 2018;35(3):175–87.
2017;51(19):1419–24. 160. Hackett G, Cole N, Saghir A, Jones P, Strange RC,
148. Maiorino MI, Bellastella G, Esposito K.  Diabetes Ramachandran S.  Testosterone replacement ther-
and sexual dysfunction: current perspectives. apy: improved sexual desire and erectile function
Diabetes Metab Syndr Obes. 2014;7:95–105. in men with type 2 diabetes following a 30-week
149. Look AHEAD Research Group, Wing RR.  Long-­ randomized placebo-controlled study. Andrology.
term effects of a lifestyle intervention on weight 2017;5(5):905–13.
and cardiovascular risk factors in individuals 161. Spitzer M, Basaria S, Travison TG, Davda MN,
with type 2 diabetes mellitus: four-year results Paley A, Cohen B, Mazer NA, Knapp PE, Hanka S,
of the Look AHEAD trial. Arch Intern Med. Lakshman KM, Ulloor J, Zhang A, Orwoll K, Eder
2010;170(17):1566–75. R, Collins L, Mohammed N, Rosen RC, DeRogatis
150. Boddi V, Castellini G, Casale H, Rastrelli G, Boni L, L, Bhasin S.  Effect of testosterone replacement on
Corona G, Maggi M.  An integrated approach with response to sildenafil citrate in men with erectile
vardenafil orodispersible tablet and cognitive behav- dysfunction: a parallel, randomized trial. Ann Intern
ioral sex therapy for treatment of erectile dysfunc- Med. 2012;157(10):681–91.
tion: a randomized controlled pilot study. Andrology. 162. Bhasin S, Brito JP, Cunningham GR, Hayes FJ,
2015;3(5):909–18. Hodis HN, Matsumoto AM, Snyder PJ, Swerdloff
151. Chen L, Staubli SE, Schneider MP, Kessels AG, Ivic RS, Wu FC, Yialamas MA.  Testosterone therapy
S, Bachmann LM, Kessler TM.  Phosphodiesterase in men with hypogonadism: an endocrine society
5 inhibitors for the treatment of erectile dysfunc- clinical practice guideline. J Clin Endocrinol Metab.
tion: a trade-off network meta-analysis. Eur Urol. 2018;103(5):1715–44.
2015;68(4):674–80. 163. Saad F, Yassin A, Almehmadi Y, Doros G, Gooren
152. Zucchi A, Costantini E, Scroppo FI, Silvani M, L.  Effects of long-term testosterone replacement
Kopa Z, Illiano E, Petrillo MG, Cari L, Nocentini therapy, with a temporary intermission, on glycemic
G.  The first-generation phosphodiesterase 5 inhibi- control of nine hypogonadal men with type 1 diabe-
tors and their pharmacokinetic issue. Andrology. tes mellitus  - a series of case reports. Aging Male.
2019;7(6):804–17. 2015;18(3):164–8.
153. Hirano T, Mori Y.  Anti-atherogenic and anti-­ 164. Padma-Nathan H, Hellstrom WJ, Kaiser FE, Labasky
inflammatory properties of glucagon-like peptide-1, RF, Lue TF, Nolten WE, Norwood PC, Peterson
glucose-dependent insulinotropic polypeptide, and CA, Shabsigh R, Tam PY, Place VA, Gesundheit
dipeptidyl peptidase-4 inhibitors in experimental N. Treatment of men with erectile dysfunction with
animals. J Diabetes Investig. 2016;7(Suppl 1):80–6. transurethral alprostadil. Medicated Urethral System
528 V. Spallone et al.

for Erection (MUSE) Study Group. N Engl J Med. 177. Althof SE, McMahon CG, Waldinger MD, Serefoglu
1997;336(1):1–7. EC, Shindel AW, Adaikan PG, Becher E, Dean
165. Heaton JP, Lording D, Liu SN, Litonjua AD, J, Giuliano F, Hellstrom WJ, Giraldi A, Glina S,
Guangwei L, Kim SC, Kim JJ, Zhi-Zhou S, Israr D, Incrocci L, Jannini E, McCabe M, Parish S, Rowland
Niazi D, Rajatanavin R, Suyono S, Benard F, Casey D, Segraves RT, Sharlip I, Torres LO. An update of
R, Brock G, Belanger A. Intracavernosal alprostadil the International Society of Sexual Medicine’s guide-
is effective for the treatment of erectile dysfunction lines for the diagnosis and treatment of premature
in diabetic men. Int J Impot Res. 2001;13(6):317–21. ejaculation (PE). J Sex Med. 2014;11(6):1392–422.
166. Moncada I, Martinez-Salamanca J, Ruiz-Castañe E, 178. Sansone A, Aversa A, Corona G, Fisher AD, Isidori
Romero J. Combination therapy for erectile dysfunc- AM, La Vignera S, Limoncin E, Maggi M, Merico
tion involving a PDE5 inhibitor and alprostadil. Int J M, Jannini EA. Management of premature ejacula-
Impot Res. 2018;30(5):203–8. tion: a clinical guideline from the Italian Society
167. Kim S, Cho MC, Cho SY, Chung H, Rajasekaran of Andrology and Sexual Medicine (SIAMS). J
MR. Novel emerging therapies for erectile dysfunc- Endocrinol Investig. 2020;44:1103.
tion. World J Mens Health. 2021;39(1):48–64. 179. McCabe MP, Sharlip ID, Lewis R, Atalla E, Balon
168. Scherzer ND, Dick B, Gabrielson AT, Alzweri LM, R, Fisher AD, Laumann E, Lee SW, Segraves
Hellstrom WJG.  Penile prosthesis complications: RT.  Incidence and prevalence of sexual dysfunc-
planning, prevention, and decision making. Sex Med tion in women and men: a consensus statement from
Rev. 2019;7(2):349–59. the Fourth International Consultation on Sexual
169. Dicks B, Bastuba M, Goldstein I.  Penile revascu- Medicine 2015. J Sex Med. 2016;13(2):144–52.
larization--contemporary update. Asian J Androl. 180. Malavige LS, Jayaratne SD, Kathriarachchi ST,
2013;15(1):5–9. Sivayogan S, Fernando DJ, Levy JC.  Erectile dys-
170. Hsieh CH, Hsu GL, Chang SJ, Yang SS, Liu SP, function among men with diabetes is strongly
Hsieh JT. Surgical niche for the treatment of erectile associated with premature ejaculation and reduced
dysfunction. Int J Urol. 2020;27(2):117–33. libido. J Sex Med. 2008;5(9):2125–34.
171. Dong L, Chang D, Zhang X, Li J, Yang F, Tan K, 181. Bellastella G, Maiorino MI, Olita L, Della Volpe E,
Yang Y, Yong S, Yu X. Effect of low-intensity extra- Giugliano D, Esposito K.  Premature ejaculation is
corporeal shock wave on the treatment of erectile associated with glycemic control in Type 1 diabetes.
dysfunction: a systematic review and meta-analysis. J Sex Med. 2015;12(1):93–9.
Am J Mens Health. 2019;13(2):1557988319846749. 182. Di Sante S, Mollaioli D, Gravina GL, Ciocca
172. Bahk JY, Jung JH, Han H, Min SK, Lee G, Limoncin E, Carosa E, Lenzi A, Jannini
YS.  Treatment of diabetic impotence with umbili- EA.  Epidemiology of delayed ejaculation. Transl
cal cord blood stem cell intracavernosal transplant: Androl Urol. 2016;5(4):541–8.
preliminary report of 7 cases. Exp Clin Transplant. 183. Sexton WJ, Jarow JP.  Effect of diabetes melli-
2010;8(2):150–60. tus upon male reproductive function. Urology.
173. Huang CY, Shih CM, Tsao NW, Lin YW, Huang PH, 1997;49(4):508–13.
Wu SC, Lee AW, Kao YT, Chang NC, Nakagami 184. Lindau ST, Tang H, Gomero A, Vable A, Huang ES,
H, Morishita R, Ou KL, Hou WC, Lin CY, Shyu Drum ML, Qato DM, Chin MH.  Sexuality among
KG, Lin FY.  Dipeptidyl peptidase-4 inhibitor middle-aged and older adults with diagnosed and
improves neovascularization by increasing circu- undiagnosed diabetes: a national, population-based
lating endothelial progenitor cells. Br J Pharmacol. study. Diabetes Care. 2010;33(10):2202–10.
2012;167(7):1506–19. 185. Agochukwu-Mmonu N, Malaeb BS, Hotaling JM,
174. Assaly R, Gorny D, Compagnie S, Mayoux E, Braffett BH, Holt SK, Dunn RL, Palmer MR, Martin
Bernabe J, Alexandre L, Giuliano F, Behr-Roussel CL, Jacobson AM, Herman WH, Wessells H. Sarma
D. The favorable effect of empagliflozin on erectile AV; and the DCCT/EDIC Research Group. Risk fac-
function in an experimental model of type 2 diabe- tors for orgasmic and concomitant erectile dysfunc-
tes. J Sex Med. 2018;15(9):1224–34. tion in men with type 1 diabetes: a cross-sectional
175. Shao N, Yu XY, Yu YM, Li BW, Pan J, Wu WH, study. Int J Impot Res. 2021;33(1):59–66.
Zhang HJ, Ma XF, Hao M, Kuang HY.  Short-term 186. Alwaal A, Breyer BN, Lue TF. Normal male sexual
combined treatment with exenatide and metformin function: emphasis on orgasm and ejaculation. Fertil
is superior to glimepiride combined metformin Steril. 2015;104(5):1051–60.
in improvement of serum testosterone levels in 187. Kempeneers P, Andrianne R, Cuddy M, Blairy
type 2 diabetic patients with obesity. Andrologia. S.  Sexual cognitions, trait anxiety, sexual anxi-
2018;50(7):e13039. ety, and distress in men with different subtypes of
176. Caparrotta TM, Greenhalgh AM, Osinski K, Gifford premature ejaculation and in their partners. J Sex
RM, Moser S, Wild SH, Reynolds RM, Webb DJ, Marital Ther. 2018;44(4):319–32.
Colhoun HM.  Sodium-glucose co-transporter 2 188. Hull EM, Lumley LA, Matuszewich L, Dominguez
inhibitors (SGLT2i) exposure and outcomes in type J, Moses J, Lorrain DS. The roles of nitric oxide in
2 diabetes: a systematic review of population-based sexual function of male rats. Neuropharmacology.
observational studies. Diabetes Ther. 2021;12:991. 1994;33(11):1499–504.
Diabetic Neuropathy: Clinical Management—Genitourinary Dysfunction in Diabetes 529

189. Gray M, Zillioux J, Khourdaji I, Smith 202. Barbagallo F, Mongioì LM, Cannarella R, La
RP. Contemporary management of ejaculatory dys- Vignera S, Condorelli RA, Calogero AE.  Sexual
function. Transl Androl Urol. 2018;7(4):686–702. dysfunction in diabetic women: an update on current
190. Althof SE, McMahon CG.  Contemporary manage- knowledge. Diabetology. 2020;1(1):11–21.
ment of disorders of male orgasm and ejaculation. 203. Maseroli E, Fanni E, Cipriani S, Scavello I,
Urology. 2016;93:9–21. Pampaloni F, Battaglia C, Fambrini M, Mannucci E,
191. Arafa M, El Tabie O. Medical treatment of retrograde Jannini EA, Maggi M, Vignozzi L. Cardiometabolic
ejaculation in diabetic patients: a hope for spontane- risk and female sexuality: focus on clitoral vascular
ous pregnancy. J Sex Med. 2008;5(1):194–8. resistance. J Sex Med. 2016;13(11):1651–61.
192. Kurbatov D, Russo GI, Galstyan GR, Rozhivanov 204. Isidori AM, Pozza C, Esposito K, Giugliano D,
R, Lepetukhin A, Dubsky S, Shwartz YG, Cimino Morano S, Vignozzi L, Corona G, Lenzi A, Jannini
S, Morgia G, Sansalone S. Correction of retrograde EA.  Development and validation of a 6-item ver-
ejaculation in patients with diabetes mellitus using sion of the female sexual function index (FSFI) as a
endourethral collagen injection: preliminary results. diagnostic tool for female sexual dysfunction. J Sex
J Sex Med. 2015;12(11):2126–9. Med. 2010;7(3):1139–46.
193. Parish SJ, Hahn SR, Goldstein SW, Giraldi A, 205. Woodard TL, Diamond MP.  Physiologic measures
Kingsberg SA, Larkin L, Minkin MJ, Brown V, of sexual function in women: a review. Fertil Steril.
Christiansen K, Hartzell-Cushanick R, Kelly-­ 2009;92(1):19–34.
Jones A, Rullo J, Sadovsky R, Faubion SS.  The 206. Caruso S, Rugolo S, Mirabella D, Intelisano G, Di
International Society for the Study of women’s Mari L, Cianci A. Changes in clitoral blood flow in
sexual health process of care for the identification of premenopausal women affected by type 1 diabetes
sexual concerns and problems in women. Mayo Clin after single 100-mg administration of sildenafil.
Proc. 2019;94(5):842–56. Urology. 2006;68(1):161–5.
194. Pontiroli AE, Cortelazzi D, Morabito A.  Female 207. Krakowsky Y, Grober ED.  A practical guide to
sexual dysfunction and diabetes: a systematic review female sexual dysfunction: an evidence-based
and meta-analysis. J Sex Med. 2013;10(4):1044–51. review for physicians in Canada. Can Urol Assoc J.
195. Rahmanian E, Salari N, Mohammadi M, Jalali 2018;12(6):211–6.
R.  Evaluation of sexual dysfunction and female 208. Wing RR, Bond DS, Gendrano IN III, Wadden
sexual dysfunction indicators in women with type T, Bahnson J, Lewis CE, Brancati F, Schneider S,
2 diabetes: a systematic review and meta-analysis. Kitabchi AE, Van Dorsten B, Rosen RC.  Sexual
Diabetol Metab Syndr. 2019;11:73. Dysfunction Subgroup of the Look AHEAD
196. Enzlin P, Rosen R, Wiegel M, Brown J, Wessells Research Group. Effect of intensive lifestyle inter-
H, Gatcomb P, Rutledge B, Chan KL, Cleary vention on sexual dysfunction in women with type
PA. DCCT/EDIC Research Group. Sexual dysfunc- 2 diabetes: results from an ancillary Look AHEAD
tion in women with type 1 diabetes: long-term find- study. Diabetes Care. 2013;36(10):2937–44.
ings from the DCCT/EDIC study cohort. Diabetes 209. Krysiak R, Drosdzol-Cop A, Skrzypulec-Plinta
Care. 2009;32(5):780–5. V, Okopień B.  Sexual functioning and depressive
197. Maiorino MI, Bellastella G, Castaldo F, Petrizzo M, symptoms in women with diabetes and prediabetes
Giugliano D, Esposito K. Sexual function in young receiving metformin therapy: a pilot study. Exp Clin
women with type 1 diabetes: the METRO study. J Endocrinol Diabetes. 2017;125(1):42–8.
Endocrinol Investig. 2017;40(2):169–77. 210. Islam RM, Bell RJ, Green S, Page MJ, Davis
198. Bąk E, Marcisz C, Krzemińska S, Dobrzyn-Matusiak SR. Safety and efficacy of testosterone for women: a
D, Foltyn A, Drosdzol-Cop A.  Relationships of systematic review and meta-analysis of randomised
sexual dysfunction with depression and accep- controlled trial data. Lancet Diabetes Endocrinol.
tance of illness in women and men with type 2 2019;7(10):754–66.
diabetes mellitus. Int J Environ Res Public Health. 211. Davis SR, Baber R, Panay N, Bitzer J, Cerdas
2017;14(9):1073. Perez S, Islam RM, Kaunitz AM, Kingsberg SA,
199. Basson R.  Clinical practice. Sexual desire and Lambrinoudaki I, Liu J, Parish SJ, Pinkerton
arousal disorders in women. N Engl J Med. J, Rymer J, Simon JA, Vignozzi L, Wierman
2006;354(14):1497–506. ME.  Global consensus position statement on the
200. Maseroli E, Scavello I, Vignozzi L. Cardiometabolic use of testosterone therapy for women. Climacteric.
risk and female sexuality-Part II.  Understanding 2019;22(5):429–34.
(and overcoming) gender differences: the key role 212. Chivers ML, Rosen RC.  Phosphodiesterase type
of an adequate methodological approach. Sex Med 5 inhibitors and female sexual response: faulty
Rev. 2018;6(4):525–34. protocols or paradigms? J Sex Med. 2010;7(2 Pt
201. Davis SR, Davison SL, Donath S, Bell RJ. Circulating 2):858–72.
androgen levels and self-reported sexual function in
women. JAMA. 2005;294(1):91–6.
Index

A B
Abnormal mitochondrial morphology, 186 Baroreflex sensitivity testing, 213
Acceptance and commitment therapy (ACT), 231, 448 BDNF-mediated spinal disinhibition, 131
Actinobacteria, 405 BDNF, see Brain-derived neurotrophic factor (BDNF)
Action to control cardiovascular risk in diabetes Behavioural therapy (BT), 231
(ACCORD), 244 Benfotiamine, 320, 321
Adrenergic receptors, 368 β common receptor (βcR) subunits, 140
Advanced glycation end-products (AGEs), 258, 286, β-hydroxybutyrate (β-OHB), 402
288, 313, 353, 389, 406 Bioenergetics, 280, 285, 286
Age-related muscle weakness, 184 “Black-box” approach, 212
Age-related sarcopenia, 188 Bladder dysfunction, 28, 491–493
Aldose reductase inhibitors (ARIs), 254, 337 diagnosis, 496, 498
Allodynia, 74 epidemiology, 492, 493
Alpha lipoic acid (ALA), 319, 444 pathophysiology, 493–494, 496
Alzheimer’s disease, 136, 381, 383 treatment, 499–501
Ambulatory BP monitoring (ABPM), 212 Blood oxygen level dependent, 431
American Academy of Neurology (AAN), 95 Brachial plexus involvement, 171
American Academy of Physical Medicine and Brain-derived neurotrophic factor (BDNF), 130, 132,
Rehabilitation (AAPM&R), 95 402, 407, 420
American Association of Electrodiagnostic Medicine Bypass angioplasty revascularization investigation in
(AAEM), 95 type 2 diabetes (BARI2D) trial, 244
American Diabetes Association (ADA), 97, 230
American Urological Association Symptom Index
(AUASI), 210 C
AMP-activated protein kinase (AMPK), 317 Calcitonin gene-related peptide (CGRP), 356
Amputation, 70 Calcium homeostasis, 132
Angiotensin receptor blockers, 264 Cardiac sympathetic imaging, 213
Angiotensin-converting enzyme (ACE), 243, 264 Cardiovascular autonomic neuropathy (CAN), 5, 21
Anterior cingulate cortex, 430 clinical care, 210, 212
Anti-inflammatory properties, 266 clinical impact and implications, 208–210
Antioxidant response element (ARE), 312 diagnosis, 210, 212, 213
Anti-viral agent, 177 diagnostic algorithm for, 211
Atypical diabetic neuropathies, 184–185 epidemiology, 203–205
Autonomic dysfunction, 4 pathophysiology, 205–207
Autonomic neuropathy, 4, 5 treatment, 213
cardiovascular autonomic neuropathy, 44 Central nervous system, 428
gastrointestinal autonomic neuropathy, 44 Charcot-Marie-Tooth disease, 244, 245
sudomotor dysfunction, 45 Chronic hyperglycemia, 185
urogenital autonomic neuropathy, 45 Clinical medicine, 4
Axo-glial interaction, 293 Clinical sensory function, 74
Axon regeneration, 339 CNTF depletion, 134
Axon terminals, 327, 341 Coenzyme Q (CoQ), 308
Axotomy, 339 Cognitive dysfunction, 381, 382

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature 531
Switzerland AG 2023
S. Tesfaye et al. (eds.), Diabetic Neuropathy, Contemporary Diabetes,
https://doi.org/10.1007/978-3-031-15613-7
532 Index

implications, 389, 391 Diabetic distal symmetric polyneuropathy (diabetic DSP)


structural brain changes, 384–386, 388 asymptomatic clinical presentation, 71, 72
type 1 diabetes, 383 clinical manifestations, 70
type 2 diabetes, 383, 384 clinical risk factors, 69
COMBO‐DPN study, 224 complications, 21
The Committee for Medicinal Products for Human Use composite symptom scale, sign scales and combined
(CHMP) of the European Medicines Agency scales, 75
(EMA), 120 diagnostic considerations, 68–69
Common diabetic cranial mononeuropathy is oculomotor differential diagnosis, 86, 88
(CNIII) mononeuropathy, 175 epidemiological studies of, 7
Compound muscle action potentials (CMAPs), 94, 177 foot complications, 69
Concurrent validity, 72 impaired protective sensation, 71
Coronary artery risk development in young adults incidence of, 19, 20
(CARDIA) study, 214 Michigan Neuropathy Screening Instrument, 76, 77
C-peptide, 136, 137 modified TCNS, 79
Cranial mononeuropathies, 175 muscle strength in, 188, 189
Cranial neuropathies, 28 neuropathic signs, 74, 75
Cyclooxygenase-2 pathway, 264 neuropathic symptoms, 73, 74
Cystopathy, 491 neuropathy symptom score, 82, 83
NIS-LL, 79
painful diabetic neuropathy, 16, 19
D prevalence of, 7, 15, 17–18
Dementia, 381, 382 revised neuropathy disability score, 83
Depression, 221 risk factors for, 20
Detection of ischemia in asymptomatic diabetics staging of, 68
(DIAD), 209 Toronto clinical neuropathy score, 77–79
DFNS protocol in type 1 and 2 diabetes, 8–14
clustering approach, 118, 120 Utah early neuropathy scale, 80, 82
of quantitative sensory testing, 114 Diabetic erectile dysfunction, 502–509, 511, 512, 514
sensory profiling, 117, 118, 122, 123 Diabetic lumbosacral radiculoplexus neuropathy
sensory testing, 121 (DLRPN), 168
thermal thresholds, 115, 116 clinical presentation, 167
Z-transformation and reference values, 116, 117 diagnosis, 169
Diabetes, 279, 282, 287, 290, 351 epidemiology, 167
Diabetes control and complications trial (DCCT), 253, historical perspective, 166
318 management, 169
Diabetes distress, 221 outcome and prognosis, 170
Diabetes mellitus, 174, 327 pathophysiology, 168, 169
Diabetes neuropathies Diabetic mononeuropathies, 28, 165
acute painful-distal sensorimotor polyneuropathies, Diabetic myopathy, 186
42 Diabetic neurons, 337
classification, 38, 39 Diabetic neuropathy, 4, 166, 354, 416, 444, 494
co-morbidities of, 41 benfotiamine, 320
diabetic peripheral neuropathy, 39, 40 central mechanisms, 419–421
differential diagnosis of, 42 chaperones, 314
epidemiology, 37, 38 clinical manifestations of, 165
hyperglycaemia induced acute painful distal early history of, 3
sensorimotor polyneuropathy, 42 genomics of
painful diabetic peripheral neuropathy, 40, 41 with Charcot-Marie-Tooth, 244, 245
treatment, 43 in human, 240, 243
Diabetes prevention program (DPP), 319 neuropathic pain in, 246
Diabetic autonomic neuropathy (DAN), 453 sodium channel variants and painful diabetic
cardiovascular autonomic neuropathy, 21 neuropathy, 246–248
incidence of, 27 transcriptional profiling, 245
prevalence of, 22–26 in type 2 diabetes patients, 241–242
risk factors for, 28 incidence of, 5
prevalence of, 25 mGluRs, 314
Diabetic cervical radiculoplexus neuropathy (DCRPN), mitochondrial dysfunction, 308–311
170 neuropathic pain, 415
Diabetic cranial neuropathies, 165 NF-kappaB, 312, 313
Index 533

nitrosative stress, 313, 318 E


oxidative stress, 310, 312 Electrochemical sweat conductance (ESC), 104
peripheral mechanisms, 416–419 Electromyography, 188
PGC-1α, 309 Endothelial dysfunction, 419
risk factors, 415 Endothelial nitric oxide synthase (eNOS), 354
SIRT1, 309 Endothelium, 353
TFAM, 310 Endothelium dysfunction, 355
Diabetic peripheral neuropathy (DPN), 51, 255, 397 Endothelium-derived hyperpolarizing factor (EDHF),
clinical features of, 53–55 261
clinical trials in, 58, 60–62 Entrapment neuropathy, 172
corneal confocal microscopy, 104–106 Epalrestat, 257
dietary interventions, 399–405 Epidemiology of Diabetes Interventions and
exercise and physical activity, 406, 407 Complications (EDIC) study, 203
high fat diet, 398, 399 Epidermal innervation, 99
microbiome, 405, 406 Epineurial arterioles, 259
nerve conduction studies, 94–97 Epo receptor (EpoR) homodimer, 140
obesity, 398 Erectile dysfunction (ED), 28, 210
pharmacologic treatment, 397, 398 Erythropoietin (Epo), 140
quantitative sensory testing, 101–103 Estimated glomerular filtration rate (eGFR), 209
risk factors for, 55, 57 Eubacterium rectale, 405
skin biopsy for IENFD measurement, 98–104 European Insulin-Dependent Diabetes Mellitus
sudomotor testing, 103–105 Prospective Complications Study
Diabetic polyneuropathy (DPN), 94, 327 (EURODIAB IDDM), 203–204
Diabetic radiculoplexus neuropathy (DRPN), 29, 165, Exercise training, 195
166
Diabetic thoracic radiculoneuropathy (DTRN), 171, 172
Diabetic truncal mononeuropathy, 171 F
Diagnostic accuracy, 78, 83 Face validity, 82, 83
Diagnostic tests, 93 Facial paralysis, 177
Diffusion tensor imaging (DTI), 388 Facial weakness, 177
Dihydrolipoic acid (DHLA) metabolism, 258 Female sexual dysfunction (FSD), 517–520
Dipeptidyl peptidase IV (DPP-IV), 264 Female sexual function index (FSFI-R), 210
Distal symmetric polyneuropathy (DSPN), 5, 51, 183, Fibular mononeuropathy, 174
221, 232 Fludrocortisone, 460
in clinical care, 230, 231 Foot complications, 70
clinical presentation, 184, 185 Foot dorsiflexion, 75
depression and anxiety, 226, 227 Functional magnetic resonance imaging (fMRI), 431
diagnostic approach, 186
generalized anxiety vs. specific fears, 227, 228
generic health status/QoL Instruments, 228, 229 G
generic to DPN-specific QoL assessments, 229, 230 GABAA receptor, 131
interventions, 194–196 Gabapentin, 444, 447
motor function, 194 Gastric emptying, 482
neurogenic muscle weakness, 186 Gastrointestinal autonomic neuropathy, 28
pathophysiology of, 185 Gastrointestinal neuropathy, 471
on physical and mental functioning, 222, 223 autonomic function, 471, 472
psychological treatment options for, 231, 232 esophagus, 472, 473
psychosocial factors, 224–226 gallbladder, 480
sleep and mental health, 224 gastric emptying, 474, 481–483
Distal vessels, 358 postprandial hypotension, 483, 484
Dorsal root ganglia (DRGs), 280, 328, 329 prokinetic agents, 479
diabetes mellitus, 331 small and large intestine, 480–482
injury, 329, 330 stomach, 473–478, 480
insulin, 337, 338 Gastroparesis, 28
sensory neurons, 335–337 GDNF family ligands, 133
Droxidopa, 461 Gene transfer, 141
Dynamic mechanic allodynia (DMA), 116, 443 Genome wide association study (GWAS) approach, 240
Dynamometry, 190 Glabrous skin, 98
Dysesthesia, 441 Glucagon-like peptide 1 (GLP-1), 338
Dyslipidemia, 279, 283, 287 Glucose transporters (GLUTs), 283
534 Index

Glutamate carboxypeptidase II, 315 M


Glutamatergic receptors, 368 Magnetic resonance imaging (MRI), 430
Glycemia, 473 Male sexual dysfunction, 502
Glycemic control, 4, 72 Manual muscle testing (MMT), 190
Grey matter, 430 Mechanical detection threshold (MDT), 116
Mechanical pain sensitivity (MPS), 116
Mechanical pain threshold (MPT), 116
H Median mononeuropathy across the wrist (MNW), 172
Heart rate (HR), 204 Mediterranean diet, 399
Heat shock proteins (HSPs), 314 Meissner’s corpuscles, 98
Hedgehog pathway agonism, 139 Mendelian disorders, 240
Hereditary sensorimotor neuropathy, 430 Metabotropic glutamate receptors (mGluRs), 314
Hexosamine pathway, 260 Michigan Neuropathy Screening Instrument (MNSI), 76,
High fat diet (HFD), 398 77
Histone deacetylases (HDACs), 405 Microcirculation, 352
Hospital Anxiety and Depression Scale, 221 Microvascular changes, 160
Hyperalgesia, 74 Microvascular disease, 351–353
Hypercholesterolemia, 185 autonomic denervation, 355, 356
Hyperglycemia, 352 diabetic neuropathy, 354, 355
Hyperglycemic rodents, 130 nerve axon reflex, 356–358
Hypertriglyceridemia, 185 Microvascular ischemia, 176
Hypogonadism, 504 Microvessel wall, 168
Midodrine, 460
Mindfulness-based cognitive therapy, 233
I Mitochondria, 307
IGF-1 receptor (IGF-1R), 137 Mitochondrial function, 366, 367
IL-6 cytokine family, 133–135 Modified TCNS (mTCNS), 79
IL-6 receptor sub-unit (IL-6R), 135 Monocarboxylic acid transporter 1 (MCT1), 402
Inflammation, 282, 363–364, 419 Mononeuropathies, 5, 28
Insulin, 4, 136 entrapment mononeuropathies, 46
Insulin-like growth factors, 137 microvasculitic diabetic mononeuropathies, 45
International Index of Erectile Function (IIEF), 210 Morphogens, 138
Intraepidermal fibers (IENFs), 99 MOS sleep problems index, 222
Intraepidermal nerve fiber density (IENFD), 93 Motor and sensory nerves, 183
Intramuscular fat-accumulations, 188–189 Motor function, 75, 194
Intravenous immunoglobulin (IVIG), 170 Motor involvement and diabetes, 196
Intravenous methylprednisolone (IVMP), 169 Motor nerve dysfunction, 187
Ipswich touch test, 75 Motor neuropathy, 183–185, 190, 191, 196
Ischemic nerve injury, 171 MTHFR gene, 243
Ischemic reperfusion injury (IR), 207 Multi-ethnic study of atherosclerosis (MESA), 204
Multiple regression analysis, 225
Muscle size, 190
J Muscle strength
JAK/STAT signaling, 134 assessment of, 189, 190
diabetic distal symmetric polyneuropathy, 188, 189
Muscle weakness, 191
K Muscular atrophy, 186, 190, 191
KCC2 expression, 131 Myopathy, 186
Ketogenic diets, 400, 401

N
L NAD-dependent sorbitol dehydrogenase, 254
Langerhans cells, 142 Native peptide sequence, 142, 143
Lateral femoral cutaneous mononeuropathy, 174 Nephropathy, 358
Linear regression models, 228 Neprilysin, 265
Lipoic acid (LA), 258 Nerve axon reflex, 356, 357
Lipoprotein lipase (LPL), 293 Nerve conduction studies (NCS), 93, 96, 183
Lumbosacral DRPN, 29 Neural impairment, 263
Index 535

Neuropad, 104 disease-modifying therapies, 297–299


Neuropathic pain, 113, 415, 429, 446 dyslipidemia, 290–293, 296
Neuropathy, 4, 358 inflammation, 282
Neuropathy symptom score, 82, 83 lipid metabolism, 283, 291, 292
Neurostimulation, 448 metabolic syndrome, 279, 281–282
Neuroticism, 225 mitochondria, 283
Neurotrophic signaling, 143 neuron, 280, 283, 285
Neurotrophic support, 127 prediabetes, 279
Neurotrophin family, 128, 494 schwann cells, 280
BDNF, 130, 132 Perivascular inflammatory, 169
GDNF family ligands, 133 Peroxynitrite, 261
IL-6 cytokine family, 133–135 Physical tests, 194
NGF, 129 Pilot longitudinal study, 78
NGF injection therapy, 129 Pinprick hyperalgesia, 116
Nitrosative stress, 313 Plantarflexion, 75
Non-contractile tissue infiltration, 186 Polyol pathway, 254, 255
Non-enzymatic glycation, 258–260 Polyradiculopathies, 5
Nuclear factor-2 erythroid related factor-2 (Nrf2), 264 Positron emission tomography (PET), 433
Postprandial hypotension, 483
Posturography, 194
O Prediction of Arrhythmic Events with Positron
Ocular motor mononeuropathies (OMM), 175, 176 Emission Tomography (PAREPET)
Olmsted County Minnesota, 175 study, 208
Omega-3 polyunsaturated fatty acids (PUFA), 264, 265, Pregabalin, 444, 447
267 Premature ejaculation (PE), 514–516
Orthostatic hypotension, 214, 453, 454 Progressive resistance training, 195
clinical presentation, 464 Proinflammatory peptides, 365
diagnostics, 465–467 Proliferator-activated receptor-gamma co-activator 1α
evaluation, 456 (PGC-1α), 309
initial steps, 455 Proof-of-concept study, 267
neurogenic supine hypertension, 462 Protein kinase C (PKC), 260, 286
nonpharmacological interventions, 458–462 Psychological flexibility, 225
postprandial hypotension, 463
sudomotor dysfunction, 463
symptoms, 455 Q
treatment, 458, 467 Quantitative sensory testing (QST), 320, 428
valsalva maneuver, 457, 458 application, in clinical trials, 102
Outer mitochondrial membrane (OMM), 284 point of care, 101, 102
Oxidative phosphorylation (OXPHOS), 289 Quantitative sudomotor axon reflex testing (QSART),
Oxidative stress, 260, 262, 282, 311 103

P R
Pain management, 113 Radiculopathies, 28
Painful diabetic polyneuropathy (P-DPN), 113, 427, 428, Radiculoplexus neuropathies, 46, 47
441 Randomized controlled trials (RCTs), 231
mononeuropathies, 441 Ranirestat, 257
non pharmacological treatment, 448 Rate-dependent depression (RDD), 131
pharmacological treatment, 444–447 Reactive oxygen species (ROS), 308, 369
radiculoplexopathy, 441 Rearranged during transfection, 133
Painful neuropathy, 364, 367 Receptor for advanced glycation end products (RAGE),
Paradoxical heat sensations (PHS), 115 258
PARP inhibitor 3-aminobenzamide, 263 Renin-angiotensin system (RAS), 264
Patient reported outcomes (PROs), 229 Reproducibility, 82
Periaqueductal grey, 431 Retinopathy, 358
Peripheral arterial disease (PAD), 358 Retrograde ejaculation, 516
Peripheral neuropathy (PN), 279, 280, 282 Rho kinase (ROCK), 367
bioenergetics, 283, 285, 288 Roux-Y gastric bypass (RYGB), 313
536 Index

S T
Sandvik Severity Index, 210 Tanezumab, 129
Saturated fatty acids (SFA), 291 Test quality and reliability, 76, 83
Scale face validity, 76, 80, 82 Thermal thresholds, 115, 116
Schwann cells, 127, 130, 133, 134, 254, 258, 280 Thermoregulatory sweat testing (TST), 104
Sensory modalities, 83 Tibial nerve stimulation, 500
Sensory nerve action potentials (SNAPs), 94 Tingling, 441
Sensory organs, 98 Toronto clinical neuropathy score (TCNS), 77–79
Serotonin, 444, 446 Toronto consensus criteria, 94
Serotonin/noradrenaline reuptake inhibitors (SSRI/ Toronto diabetic neuropathy expert consensus, 243
SNRI), 370 Transient receptor potential (TRP), 368
Severe acute respiratory syndrome coronavirus 2 Treatment induced neuropathy (TIN), 29
(SARS-CoV-2) pandemic, 86 Treatment induced neuropathy of diabetes (TIND)
Severity, 71 clinical features of, 158
Short Form (SF)-36, 224 historical background, 157, 158
Short-form McGill Pain Questionnaire (SF-MPQ-2), 231 Tricarboxylic acid (TCA) cycle, 207
Silent mating type information regulation 2 homolog1, Tricyclic antidepressants (TCAs), 444, 445
309 Tumor necrosis factor alpha (TNFα), 366
Single nucleotide polymorphisms (SNPs), 243
Skin biopsy, 99, 100, 188
Sleep problem index, 222 U
Small and large fiber function, 94 Ulnar mononeuropathy across the elbow (UNE), 173
Small fiber dysfunction, 74 United Kingdom Prospective Diabetes Study (UKPDS),
Small vessel diseases (SVDs), 385 253, 352
Small-fiber neuropathy (SFN), 93 Urethra dysfunction, 495
Smooth muscle actin (SMACTIN) staining, 169 Urinary incontinence, 28
Sonic hedgehog protein (Shh), 340 Urothelial dysfunction, 494
Sorbinil, 255 Utah early neuropathy scale (UENS)
Sorbitol dehydrogenase (SORD) gene, 244 examination, 80–82
Spinal cord, diabetic neuropathy, 363
inflammation, 364, 366
molecular mechanisms, 366, 367 V
proinflammatory peptides, 365, 366 Valsalva maneuver, 456
Spinal cord stimulation (SCS), 373, 374, 448 Vascular disease, 351
Spinal disinhibition, 370–373 Vascular endothelial growth factor (VEGF), 139, 140
Standard ENG examination, 187 Vasopeptidase inhibitors, 265
Stem cells and secretomes, 143, 144 Visual Analog Scale (VAS), 443
Sudomotor testing, 453, 456 Voltage-gated calcium channels, 369
electrochemical sweat conductance, 104 Voltage-gated sodium channels, 369
neuropad, 104
quantitative sudomotor axon reflex testing, 103
thermoregulatory sweat testing, 104 W
Superoxide, 261 Ward, John D, 4
Survival motor neuron (SMN), 336 White matter, 430
Symmetric polyneuropathy, 5 White matter hyperintensities (WMH), 386
Sympathetic skin response (SSR), 466 Wide dynamic range (WDR), 367
Synaptic plasticity, 367 Williamson, RT, 4

You might also like