You are on page 1of 5

Clinical Practice: Mini-Review

Nephron 2019;143:12–16 Received: June 29, 2018


Accepted after revision: August 10, 2018
DOI: 10.1159/000493278 Published online: October 1, 2018

Inflammation in Diabetic Kidney Disease


Rosa E. Pérez-Morales a María Dolores del Pino b José Manuel Valdivielso c, d
     

Alberto Ortiz d–f Carmen Mora-Fernández d, f, g Juan F. Navarro-González a, d, f, g


     

a Servicio
de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain;
b Servicio
de Nefrología, Hospital Torrecárdenas, Almería, Spain; c Grupo de Investigación Traslacional Vascular y
 

Renal, Instituto de Investigación Biomédica IRBLLEIDA, Lleida, Spain; d Red de Investigación Renal (REDINREN),
 

Instituto de Salud Carlos III, Madrid, Spain; e Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-
 

Universidad Autónoma de Madrid y Fundación Renal Iñigo Álvarez de Toledo-Instituto Reina Sofía de Investigación
Nefrológica, Madrid, Spain; f GEENDIAB (Grupo Español para el Estudio de la Nefropatía Diabética), Sociedad
 

Española de Nefrología, Spain; g Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa
 

Cruz de Tenerife, Spain

Keywords Introduction
Diabetes · Diabetic kidney disease · Inflammation ·
Therapies targeting inflammation Diabetic kidney disease (DKD) is the most prevalent
chronic renal disease and the major cause of end-stage
renal failure worldwide, predominantly due to the grow-
Abstract ing incidence of Type 2 diabetes associated with obesity
Background: Diabetes is a growing public health problem. [1]. DKD has been classically considered the consequence
Diabetic kidney disease (DKD) is the most prevalent chronic from the interaction between hemodynamic and meta-
renal disease and the major cause of end-stage renal failure bolic factors, but current knowledge indicates that its
worldwide, predominantly due to the increase of Type 2 dia- pathogenesis is multifactorial, where the immune re-
betes associated with obesity. The intimate mechanisms sponse and inflammation play a major role [2, 3]. In ad-
leading to the development and progression of renal injury dition, pro-inflammatory signaling pathways and their
in DKD are not well understood, but current knowledge in- downstream products are emerging as new biomarkers
dicates that its pathogenesis is multifactorial, where the im- and promising therapeutic targets [4].
mune response and inflammation appear to be relevant fac-
tors. Summary: This review summarizes the role of relevant
inflammatory molecules and pathways that participate in Inflammation in DKD
the development of DKD. Likewise, we focused on the new
therapeutic approaches based on anti-inflammatory effects Diabetes mellitus (DM) is associated with hemody-
of different drugs. Key Messages: This new pathogenic per- namic and metabolic alterations that produce the activa-
spective of DKD as an inflammatory condition leads to novel tion of diverse transduction pathways in virtually all types
horizons, such as the potential role of inflammatory signal-
ing pathways and their downstream products as emerging
biomarkers and promising therapeutic targets. Contribution from the CME Course of the DIABESITY Working Group
© 2018 S. Karger AG, Basel of the ERA-EDTA, Lisbon, November 24–25, 2017.

© 2018 S. Karger AG, Basel Dr. Juan F. Navarro-González


Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria
Carretera del Rosario 145
E-Mail karger@karger.com
ES–38010 Santa Cruz de Tenerife (Spain)
www.karger.com/nef E-Mail jnavgon @ gobiernodecanarias.org
of kidney cells [2, 3]. DKD is associated with both sys- facilitating neutrophil infiltration of the tubule-intersti-
temic and local renal inflammation with the participation tium, influences extracellular matrix dynamics, and pro-
of crucial inflammatory cells, molecules, and pathways, motes overall kidney hypertrophy, thickening of the glo-
such as macrophages, the nuclear transcription factor- merular basement membrane, podocyte hypertrophy,
kappa B (NFκB), the janus kinase/signal transducers and and cell cycle arrest, which is correlated with albumin-
activators of transcription (JAK/STAT) pathway, and in- uria. IL-18 stimulates the release of interferon gamma
flammatory cytokines [2, 3]. and other cytokines, increases the expression of adhesion
The mononuclear phagocyte system is activated in molecules, and induces endothelial cell apoptosis. High
DM. Macrophages infiltrate the kidney, and the cycle of serum levels of IL-18 have been noted in patients with
cytokine release and monocyte and macrophage recruit- macroalbuminuria, suggesting a role in the development
ment culminates in inflammatory-related structural of microvascular kidney complications. Early in the
changes. Other cells such as mast cells also infiltrate the course of diabetes, both glomerular and tubular cells
tubule-interstitium and releases inflammatory mediators show increased TNFα mRNA expression levels. TNF-α
and proteolytic enzymes. The magnitude of macrophage has multiple actions: induction and differentiation of in-
infiltration and the extent of mast cell degranulation is flammatory cells, cytotoxicity to kidney cells, activation
associated with the rate of loss of estimated glomerular of apoptosis, altered glomerular hemodynamics, in-
filtration (eGFR) [4]. creased vascular endothelial permeability, and increased
NFκB, a transcription factor that is activated by cyto- oxidative stress. TNFα exerts its biological actions via the
kines and oxygen radicals, controls the expression of interaction with 2 cell surface receptors, TNFα receptor 1
genes involved in different processes, such as the immune (TNFR1) and 2 (TNFR2), which have shown a promise
response and inflammation. In addition, NFκB is central value as new prognostic DKD biomarkers [7].
in the interplay among the different factors, molecules,
and pathways resulting in structural alterations and func- Chemokines
tional abnormalities observed in DKD, such as activation Chemokines are a subgroup of cytokines that function
of the renin-angiotensin system, advanced glycation end- as “chemoattractant” molecules, playing a key role in in-
products accumulation, and nicotinamide adenine dinu- flammatory cell recruitment, migration, and interaction, as
cleotide phosphate hydrogen (NADPH)-dependent oxi- well as in cellular adhesion, differentiation, and tissue dam-
dative stress [2, 3]. age in the setting of DKD [8]. Several inflammatory chemo-
kines, which are upregulated in response to metabolic and
Inflammatory Cytokines hemodynamic features of the diabetic milieu, participate in
Cytokines are a group of polypeptide signaling mole- the pathogenesis of renal damage in diabetes, particularly
cules that promote autocrine, paracrine, and juxtacrine monocyte chemotactic protein-1 (MCP-1)/chemokine
signaling as a part of the innate immune response. Pro- C-C motifligand 2 (CCL2), C-X3-C motif chemokine (CX-
duction of cytokines is induced by numerous stimuli in 3CL1), and CCL5/RANTES (C-C motif ligand 5/regulated
DM [4]. In the kidney, both blood-borne cells as well as on activation, normal T cell expressed and secreted).
diverse intrinsic renal cells (glomerular, endothelial, tu- Elevated levels of MCP-1/CCL2 have been reported in
bular, and mesangial cells) are able to synthesize inflam- biopsied kidneys and urine from patients with DKD, and
matory cytokines. The levels of these substances increase they play a role in macrophage infiltration of the tubule-
as nephropathy progresses, with an independent rela- interstitium. Angiotensin II directly induces MCP-1/
tionship between these inflammatory parameters and CCL2 expression, and blockade of the renin-angiotensin
urinary albumin excretion, suggesting a role in the patho- system (RAS) leads to the reduction of MCP-1/CCL2 in
genesis of DKD [2, 3]. urine. Other biological roles for MCP-1/CCL2 include ef-
Different actions from diverse cytokines, such as inter- facement of podocyte foot processes, podocytopenia with
leukin (IL)-1, IL-6, IL-18, and tumor necrosis factor-al- glomerular basement membrane denudement, and dam-
pha (TNFα) have been involved in the pathogenesis of age to the slit diaphragm, all of which explain the correla-
DKD [5, 6]. IL-1 stimulates the production of prostaglan- tion between urine levels of MCP-1/CCL2 and albumin-
din E and the release of phospholipase A2, participating uria. The receptor for this chemokine (MCP-1/CCL2C-C
in the development of intraglomerular hemodynamic ab- chemokine receptor type 2 [CCR2]) is distributed in the
normalities. IL-1 activity is also linked to increased per- kidney mononuclear phagocyte system and possibly in
meability of vascular endothelial cells. IL-6 plays a role in differentiated podocytes. CX3CL1 and CCL5/RANTES

Inflammation in DKD Nephron 2019;143:12–16 13


DOI: 10.1159/000493278
are upregulated in diabetes, both within the glomerular including diabetes. Moreover, the activation of JAK/
and tubular cells, and in peritubular capillaries, acting as STAT is an important mechanism by which hyperglyce-
chemoattractants for immune cells and for cellular adhe- mia contributes to renal damage [11].
sion.
NADH Oxidase and Suppressors of Cytokine Signaling
Adhesion Molecules Chronic hyperglycemia in combination with growth
Intercellular adhesion molecule 1 (ICAM-1), vascular factors and cytokines act on cells to impair redox balance
cell adhesion protein 1, endothelial cell-selective adhe- due to either increased generation of ROS or insufficient
sión molecule, and E-selectin are cell surface-localized antioxidant defense systems, thus resulting in oxidative
factors that facilitate intercellular binding, adhesion, and damage of biological macromolecules (lipids, proteins,
intercellular communication, participating in the patho- and DNA) and tissue injury. Potential sources of ROS in-
genesis of DKD [9]. The expression of adhesion mole- clude NADPH oxidase (Nox), a multi-subunit enzyme
cules in DKD is increased in response to TNFα, NFκB, that catalyzes the generation of the reactive-free radical
and hemodynamic shear stress. ICAM-1 exhibits high ex- superoxide by reduction of O2 using either NADPH or
pression in resident kidney cells, and elevated urine levels NADH as a substrate. Collectively known as the Nox fam-
of ICAM-1 have been related to DKD progression. High ily, 7 isoforms are expressed in mammals, Nox1 and Nox4
circulating levels of soluble forms of vascular cell adhe- being the main isoforms involved in DKD [12].
sion protein 1 and ICAM-1 are also related to the progres-
sion of DKD from microalbuminuria to macroalbumin-
uria. Endothelial cell-selective adhesion molecule reduces Therapies Targeting Inflammation: Future
glomerular permeability, and its downregulation in early Perspectives
DKD may promote albuminuria, whereas soluble levels
of E-selectin are positively correlated with albuminuria New therapeutic strategies in DKD have moved to-
and with cardiovascular disease. ward anti-inflammatory agents as potential new treat-
ments [13] (Table 1).
Inflammatory Signal Transduction Pentoxifylline (PTF) is a methylxanthine-derived phos-
JAK/STAT is an intracellular cytokine-associated sig- phodiesterase inhibitor with beneficial effects on microcir-
naling pathway that serves as a key mediator between culatory blood flow. A meta-analysis published in 2008 re-
paracrine stimuli and nuclear receptors. Cytokines and ported a substantial antiproteinuric effect of PTF in patients
diabetic factors activate this essential mechanism that reg- with diabetic nephropathy, and pointed to the reduction of
ulates cell activation, proliferation, recruitment, migra- proinflammatory cytokines as the most likely explanation
tion, and differentiation [10]. The upregulation of JAK/ for this action [14]. The Pentoxifylline for Renoprotection
STAT has been reported in the glomerular cells of patients in Diabetic Nephropathy study [15] was a clinical trial that
with early DKD, and tubule-interstitial expression of var- evaluated the effect of PTF in DKD patients with residual
ious JAK and STAT isoforms increases with disease pro- albuminuria despite RAS inhibitor treatment. After 24
gression and correlates inversely with the eGFR. months, the eGFR decreased by 2.1 ± 0.4 mL/min/1.73 m2
NF-kB, a key transcription factor for inflammatory in the treatment group, a significant difference (p < 0.001)
processes in the diabetic kidney, is activated via JAK/ compared with 6.5 ± 0.4 mL/min/1.73 m2 decline in the
STAT. In resident kidney cells, NF-kB is rapidly activated control group. The between-group difference in the change
by diverse stimuli, including hyperglycemia, advanced in albuminuria between the groups was also significant (5.7
glycation end-products, mechanical stress, reactive oxy- vs. –14.9%; p = 0.001). Interestingly, a recent post hoc anal-
gen species (ROS), inflammatory cytokines, angiotensin ysis of the Pentoxifylline for Renoprotection in Diabetic
II, and albuminuria/proteinuria. Upon activation, NF-kB Nephropathy trial has shown that treatment with PTF leads
stimulates the transcription of proinflammatory cyto- to a significant increase in serum and urinary Klotho con-
kines, chemokines, and adhesion molecules [6]. centrations, clinical findings supported by experimental
JAK/STAT members are controlled in a classical neg- data that show that PTF prevents the downregulation of
ative-feedback loop by the suppressors of cytokine signal- Klotho protein and mRNA expression induced by albumin
ing (SOCS) family (SOCS1–7 and CIS). The dysregulated and inflammatory cytokines in renal tubular cells [16].
JAK/STAT/SOCS pathway contributes to the pathogen- Emapticap pegol is an MCP-1/CC2 antagonist evalu-
esis of cancer, autoimmune and inflammatory disorders ated in a phase 2, placebo-controlled trial of DKD pa-

14 Nephron 2019;143:12–16 Pérez-Morales et al.


DOI: 10.1159/000493278
Table 1. Potential therapies for diabetic kidney disease targeting inflammatory pathways

Drug Target Identifier Study population Outcomes

Pentoxifylline Inflammatory EudraCT number Type 2 diabetes Mean difference in UAE of 21%
Cytokines 2007-005985-10 eGFR 15–60 (p < 0.001) and eGFR decline
UAE >30 mg/24 h 4.3 mL/min/1.73 m2 lower than in
the placebo group (p < 0.001)
Baricitinib JAK1/JAK2 NCT01683409 Type 2 diabetes Albuminuria reduction by 40%
Macroalbuminuria No effect on eGFR
eGFR 20–75 mL/min/1.73 m2
Emanticap Pegol CCL2 NCT01547897 Type 2 diabetes Albuminuria reduction by 29%
(NOX-E36) eGFR >25 mL/min/1.73 m2 compared with baseline ( p < 0.05),
UACR >100 mg/g but no significant difference with
­placebo
CCX 140-B CCR2 NCT01447147 Type 2 diabetes 18% reduction of albuminuria
eGFR ≥25 mL/min/1.73 m2 compared with placebo (p < 0.0004)
UACR 100–3,000 mg/g in the 5 mg group. No reduction
of albuminuria in the 10 mg group
CTP-499 PDE NCT01487109 Type 2 diabetes 16% UACR reduction
eGFR no limit
UACR 300–5,000 mg/g
LY3016859 TGF-α/epiregulin NCT01774981 eGFR <90 mL/min/1.73 m2 Study ongoing.
UACR >400 mg/g No results available

eGFR, estimated glomerular filtration rate; TGF-α, transforming growth factor alpha; UACR, urinary albumin-to-creatinine ratio.

tients with residual macroalbuminuria while on RAS in- by 41% at week 24 compared with placebo; there was no
hibitor therapy. After 12 weeks, urinary albumin-to-cre- change in eGFR but decreased serum levels of inflamma-
atinine ratio (UACR) was lower by 29% compared with tory biomarkers including urine C-X-C motif chemo-
baseline, without differences from the placebo group. The kine-10 and urine C-C motif ligand-2, plasma soluble
maximum difference, that is, 26% (p = 0.06) between TNFR 1 and 2, ICAM-1 and serum amyloid A [19].
emapticap and placebo, was seen 8 weeks after discon- Studies have shown that the overexpression of SOCS
tinuation of treatment [17]. in the kidney can relieve the progression of DN by inhib-
CCX140-B is a selective inhibitor of CCR2 that was iting the JAK/STAT pathway. Moreover, SOCS can also
tested in a double-blind, placebo controlled trial also en- suppress renal tubular epithelial-mesenchymal transition
rolling DKD patients with proteinuria, eGFR of 25 mL/ that is induced by oncostatin-M, a multifunctional mem-
min/1.72 m2 or higher, and managed on RAS inhibitors, ber of the IL-6 cytokine family. Finally, renal delivery of
who were treated with 5- or 10-mg doses of CCX140-B. SOCS1 and SOCS3 ameliorates proteinuria and leads to
Only participants who had uninterrupted treatment for reduced oncostatin expression and extracellular matrix
52 weeks were included in the intention-to-treat analysis. deposition [11, 20].
Treatment with the 5-mg dose resulted in a significant
decrease in albuminuria of 18% as compared to the pla-
cebo group. The albuminuria-lowering treatment effect Conclusions
persisted throughout the 52 weeks of study [18].
Baricitinib, a JAK 1/2 inhibitor, reduced UACR in a DM is a major global health problem and DKD is one
dose-dependent manner after 3 and 6 months of treat- of its most important complications. Conventional treat-
ment in a phase 2 clinical trial in DKD patients with re- ments provide incomplete kidney protection and new
sidual macroalbuminuria on RAS inhibitor treatment. A therapeutic approaches are needed. Nowadays, inflam-
daily dose of 4 mg Baricitinib decreased morning UACR mation is acknowledged as a key factor in the develop-

Inflammation in DKD Nephron 2019;143:12–16 15


DOI: 10.1159/000493278
ment and progression of DKD, and therefore, anti-in- to JFNG, and FIS PI15/00298, CP14/00133 and PI16/02057;
flammatory targets directed at specific molecular signa- ISCIII-RETIC REDinREN RD16/0009; Sociedad Española de Ne-
frología (S.E.N.) and ACINEF. We acknowledge cofounding by
tures can be promising therapeutic strategies for DKD. Fondo Europeo de Desarrollo Regional (FEDER), Unión Europea
(“Una forma de hacer Europa”).

Acknowledgments

Research activity of the authors have been supported by Insti- Disclosure Statement
tuto de Salud Carlos III (ISCIII; Spanish Ministry of Economy,
Industry and Competitiveness): EC07/90021 and FIS PI16/00024 The authors have no conflicts of interest to declare.

References
 1 Martínez-Castelao A, Navarro-González J,   8 Ruster C, Wolf G: The role of chemokines and nal function and urinary albumin excretion in
Górriz JL, de Alvaro F: The concept and the chemokine receptors in diabetic nephropa- patients with diabetic kidney disease: the
epidemiology of diabetic nephropathy have thy. Front Biosci 2008;13:944–955. PREDIAN trial. J Am Soc Nephrol 2015; 26:
changed in recent years. J Clin Med 2015; 4:   9 Avci E, Uzeli S: The role of adhesion mole- 220–229.
1207–1216. cules and cytokines in patients with diabetic 16 Navarro-González JF, Sánchez-Niño MD,
 2 García-García PM, Getino-Melián MA, nephropathy. Biomed Res 2016:S343–S348. Donate-Correa J, Martín-Núñez E, Ferri C,
Dominguez-Pimentel V, Navarro-González 10 Marrero MB, Banes-Berceli AK, Stern DM, Pérez-Delgado N, et al: Effects of pentoxifyl-
JF: Inflammation in diabetic kidney disease. Eaton DC: Role of the JAK/STAT signaling line on soluble Klotho concentrations and
Worl J Diabetes 2014;5:431–443. pathway in diabetic nephropathy. Am J Physi- renal tubular cell expression in diabetic kid-
 3 Donate-Correa J, Martín-Nuñez E, Muros- ol Renal Physiol 2006;290:762–768. ney disease. Diabetes Care 2018; 41: 1817–
de-Fuentes M, Mora-Fernández C, Navarro- 11 Ortiz-Muñoz G, Lopez-Parra V, Lopez-Fran- 1820.
González JF: Inflammatory cytokines in dia- co O, Fernandez-Vizarra P, Mallavia B, Flores 17 Menne J, Eulberg D, Beyer D, Baumann M,
betic nephropathy. J Diabetes Res 2015;2015: C, et al: Suppressors of cytokine signaling ab- Saudek F, Valkusz Z, et al: C-C motif-ligand 2
948417. rogate diabetic nephropathy. J Am Soc inhibition with emapticap pegol (NOX-E36)
  4 Alicic RZ, Johnson EJ, Tuttle KR: Inflamma- Nephrol 2010;21:763–772. in type 2 diabetic patients with albuminuria.
tory mechanisms as new biomarkers and ther- 12 Jha JC, Banal C, Chow BS, Cooper ME, Jande- Nephrol Dial Transplant 2017;32:307–315.
apeutic targets for diabetic kidney disease. leit-Dahm K: Diabetes and kidney disease: 18 de Zeeuw D, Bekker P, Henkel E, Hasslacher
Adv Chronic Kidney Dis 2018;25:181–191. role of oxidative stress. Antioxid Redox Signal C, Gouni-Berthold I, Mehling H, et al: The ef-
  5 Navarro-González J, Mora-Fernández C: The 2016;25:657–684. fect of CCR2 inhibitor CCX140-B on residual
role of inflammatory cytokines in diabetic ne- 13 Pérez-Gomez MV, Sanchez-Niño MD, Sanz albuminuria in patients with type 2 diabetes
phropathy. J Am Soc Nephrol 2008; 19: 433– AB, Zheng B, Martín-Cleary C, Ruiz-Ortega and nephropathy: a randomised trial. Lancet
442. M, et al: Targeting inflammation in diabetic Diabetes Endocrinol 2015;3:687–696.
  6 Navarro-González J, Mora-Fernández C, Mu- kidney disease: early clinical trials. Expert 19 Tuttle KR, Brosius FC 3rd, Adler SG, Kretzler
ros de Fuentes M, García-Pérez J: Inflamma- Opin Investig Drugs 2016;25:1045–1058. M, Mehta RL, Tumlin JA, et al: JAK1/JAK2
tory molecules and pathways in the pathogen- 14 McCormick B, Sydor A, Akbari A, Fergusson inhibition by baricitinib in diabetic kidney
esis of diabetic nephropathy. Nat Rev Nephrol D, Doucette S, Knoll G: The effect of pentoxi- disease: results from a phase 2 randomized
2011;7:327–340. fylline on proteinuria in diabetic kidney dis- controlled clinical trial. Nephrol Dial Trans-
  7 Hojs R, Ekart R, Bevc S, Hojs N: Markers of ease: a meta-analysis. Am J Kidney Dis 2008; plant 2018, Epub ahead of print.
inflammation and oxidative stress in the de- 52:454–463. 20 Liu Q, Xing L, Wang L, Yao F, Liu S, Hao J, et
velopment and progression of renal disease in 15 Navarro-González JF, Mora-Fernández C, al: Therapeutic effects of suppressors of cyto-
diabetic patients. Nephron 2016; 133: 159– Muros de Fuentes M, Chahin J, Méndez ML, kine signaling in diabetic nephropathy. J His-
162. Gallego E, et al: Effect of pentoxifylline on re- tochem Cytochem 2014;62:119–128.

16 Nephron 2019;143:12–16 Pérez-Morales et al.


DOI: 10.1159/000493278

You might also like