You are on page 1of 14

Journal of the Formosan Medical Association (2018) 117, 662e675

Available online at www.sciencedirect.com

ScienceDirect

journal homepage: www.jfma-online.com

Review Article

Update of pathophysiology and management


of diabetic kidney disease
Yi-Chih Lin a, Yu-Hsing Chang b, Shao-Yu Yang b,*,
Kwan-Dun Wu b, Tzong-Shinn Chu b

a
Department of Medicine, National Taiwan University Hospital Jinshan Branch, New Taipei City,
Taiwan
b
Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan

Received 5 November 2017; received in revised form 5 February 2018; accepted 8 February 2018

KEYWORDS Diabetic kidney disease (DKD) is a major cause of morbidity and mortality in patients with dia-
Diabetic kidney betes mellitus and the leading cause of end-stage renal disease in the world. The most char-
disease (DKD); acteristic marker of DKD is albuminuria, which is associated with renal disease progression and
Diabetic nephropathy cardiovascular events. Renal hemodynamics changes, oxidative stress, inflammation, hypoxia
(DN); and overactive renin-angiotensin-aldosterone system (RAAS) are involved in the pathogenesis
Diabetes mellitus; of DKD, and renal fibrosis plays the key role. Intensified multifactorial interventions, including
Albuminuria RAAS blockades, blood pressure and glucose control, and quitting smoking, help to prevent DKD
development and progression. In recent years, novel agents are applied for preventing DKD
development and progression, including new types of glucose-lowering agents, pentoxifylline,
vitamin D analog paricalcitol, pyridoxamine, ruboxistaurin, soludexide, Janus kinase inhibitors
and nonsteroidal minerocorticoid receptor antagonists. In this review, recent large studies
about DKD are also summarized.
Copyright ª 2018, Formosan Medical Association. Published by Elsevier Taiwan LLC. This is an
open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).

Introduction adequate glycemic control, and is one of the main causes of


death in these patients.1 DKD is defined according to the
Diabetic kidney disease (DKD), also known as diabetic ne- changes in renal structure and function. Major renal
phropathy (DN) in the past, usually occurs in patients with structural changes in DKD include mesangial expansion,
type 1 and type 2 diabetes mellitus (DM) without long-term glomerular and tubular basement membrane thickening,

* Corresponding author. Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 100,
Taiwan. Fax: þ886 2 23934176.
E-mail address: yangsy@ntuh.gov.tw (S.-Y. Yang).

https://doi.org/10.1016/j.jfma.2018.02.007
0929-6646/Copyright ª 2018, Formosan Medical Association. Published by Elsevier Taiwan LLC. This is an open access article under the CC
BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Update of pathophysiology and management of diabetic kidney disease 663

and glomerular sclerosis. DKD usually manifests a clinical processes and overactive renin-angiotensin-aldosterone
syndrome including persistent albuminuria (Table 1), system (RAAS)7 (Fig. 2).
increased blood pressure, sustained reduction in glomerular
filtration rate (GFR), increased cardiovascular events and Renal hemodynamic changes
cardiovascular event-associated mortality. Glomerular hyperfiltration leads to the occurrence of DKD.
According to the data from the International Diabetes Hyperglycemia causes afferent arteriolar dilatation by
Foundation in 2014, there were approximate 380 million release of vasoactive mediators, such as insulin-like growth
people with diabetes who accounted for 8.3% of the world factor 1 (IGF-1), glucagon, nitric oxide (NO), vascular
population. Among the causes of ESRD in the world, DM endothelial growth factor (VEGF) and prostaglandin. On the
accounted for 30e47%. In the United States, about 54.4% of other hand, alterations in renal tubular function also occur
patients with type 1 diabetes mellitus will eventually in the early stage of DM, and are related to the degree of
receive renal replacement treatment (RRT).2 According to glycemic control. Due to high filtrated load of glucose,
the data from the Bureau of Health Promotion Annual reabsorption of both glucose and sodium chloride is
Report in Taiwan in 2011, the prevalence of hyperglycemia increased because of upregulation of sodium glucose
was 9.5% and the prevalence of kidney disease in patients cotransporter 2 (SGLT2) in the proximal tubules. Thus, the
with DM was 15.42%.3 Annual Report on Kidney Disease in delivery of sodium chloride to the macula densa of distal
Taiwan in 2014 showed that the annual patient number of tubules is decreased, and then causes dilatation of afferent
incident dialysis was 10,486 in 2012, and the ratio of pa- arteriole because of tubuloglomerular feedback. At the
tients with DM in incident dialytic patients gradually same time, constriction of efferent arteriole occurs due to
increased from 36.9% in 2000 to 45.2% in 2012. Besides, the high local level of angiotensin II, and then causes changes
prevalent number of dialytic patients with DM in Taiwan of autoregulation and glomerular hypertension.8
also increased from 3778 persons (11.9% of prevalent dia- On the other hand, hyperglycemia, insulin resistance
lytic patients) in 2000 to 23,139 persons (34.2% of prevalent and compensatory hyperinsulinemia independently cause
dialytic patients) in 2012.4 The most importance of all, the endothelial dysfunction by promoting some intracellular
dialytic patients with diabetes have higher mortality than mechanisms, such as increased reactive oxygen species
those without diabetes. (ROS) production, activation of protein kinase C (PKCs) and
Albuminuria is one of the most characteristic clinical advanced glycation end-products (AGE)-induced pro-
signs in DKD. In the past, especially from the observations in inflammatory signaling. The interactions of mediators pro-
patients with type 1 DM, the clinical stages of DKD were duced by endothelial cells are disrupted and tend to
considered to begin from early glomerular hyperfiltration, imbalance. Within these mediators, endothelin-1 (ET-1) is
followed by the development of microalbuminuria, mac- the most potent vasoactive peptide produced by endothe-
roalbuminuria, and then declined GFR (Fig. 1).5 However, in lial cells in regulation of vascular homeostasis. In the
recent studies of type 2 DM, many patients with DKD do not endothelium, compensatory hyperinsulinemia increases ET-
manifest above classic step-by-step changes. Therefore, 1 secretion, thus results in vasoconstriction and vascular
the concept of natural history of DKD is changing and dysfunction.9 In the kidney, activation of endothelin-
continuing to be evolved. Albuminuria is now considered an receptor A is not only associated with vasoconstriction,
active and deteriorating condition rather than a sequent but also podocyte injury, oxidative stress, inflammation,
process in DKD.6 Development of macroalbuminuria often and fibrosis.10
accompanies with the decline of GFR and may progress to
end-stage renal disease (ESRD).2 This review summarizes Ischemia and inflammation
the recent evolution of pathophysiology and novel treat- Glomerular and vascular lesions in DKD reduce the oxygen
ment of DKD and expects providing help for clinical man- supply, cause renal medulla hypoxia and renal tubular
agement of DKD. dysfunction. In advanced DKD, multiple mechanisms reduce
the intrarenal vasodilative NO production.11 For compen-
Pathophysiology sation, the oxygen demand of the residual nephrons is
increased. Without adequate oxygen supply, more free
radicals would be produced and thus destruct renal tissues.
Conventional pathogenesis Hypoxia-inducible factor (HIF) is increased to deal with
hypoxic states. However, hyperglycemia may interfere the
Renal fibrosis, the final common pathway in the patho- stability of HIF and facilitate tissue fibrosis. The higher is
physiology of DKD, is caused by at least renal hemodynamic the blood sugar level, the stronger response is the tissue
changes, ischemia and glucose metabolism abnormalities- damage.12
associated oxidative stress increases, inflammatory

Table 1 Definition of the albuminuria in diabetic kidney disease.


Normoalbuminuria Daily urine albumin amount: <30 mg
Moderately increased albuminuria (Microalbuminuria) Daily urine albumin amount: 30 mge300 mg or
Ratio of urine albumin over urine creatinine: 30e300
Severely increased albuminuria (Macroalbuminuria) Daily urine albumin amount: >300 mg or
Ratio of urine albumin over urine creatinine: >300
664 Y.-C. Lin et al.

Figure 1 Nature history and renal changes in type 1 diabetes mellitus. Type 2 diabetes mellitus may not follow this time
course. DN, diabetic nephropathy; ESRD, end-stage renal disease; GBM, glomerular basement membrane; GFR, glomerular
filtration rate; BP, blood pressure.

Figure 2 Conventional pathophysiology and novel medical treatment of diabetes kidney disease. IGF-1, insulin-like growth
factor 1; TGF-b1, transforming growth factor b1; VEGF, vascular endothelial growth factor; NO, Nitric oxide; PG, prostaglandin; AT
II, angiotensin II; ET-1, endothelin-1; SGLT2, sodium glucose co-transporters 2; ROS, reactive oxygen species; AGEs, advanced
glycation end products; TNF, tumor necrosis factor; NF-kB, nuclear factor kappa-light-chain-enhancer of activated B cells; HIF,
hypoxia-inducible factor; RAAS, renin-angiotensin-aldosterone system; ECM, extracellular matrix; PTF, pentoxifylline; VitD,
vitamin D.

Recent researches reveal that the prevalence of Toxic metabolites are also related to renal tissue dam-
obstructive sleep apnea (OSA) among the patients with dia- age. Hyperglycemia causes mitochondrial overload and
betes mellitus is relatively high.13 OSA has been indicated to then leads to produce ROS, which further destruct
influence the control of blood sugar and blood pressure.14,15 glomerular podocytes and promote their apoptosis. AGEs
Besides, OSA and accompanying intermittent hypoxia cause also result in the protein dysfunction, and impair the
the similar pathogenesis associated with diabetic vascular metabolism of extracellular matrix.19 Moreover, smoking is
abnormalities, including activation of inflammatory also considered to cause renal damage and progression of
response and ROS reaction and production of AGEs. Treat- DKD in both type 1 and type 2 diabetes via some patho-
ment and control of OSA have potential benefit to prevent genesis, including elevated oxidative stress, upregulation
from vascular complications of diabetes mellitus.16e18 of growth factors, and endothelial dysfunction.20
Update of pathophysiology and management of diabetic kidney disease 665

Cellular and molecular experiments show that inflam- from studies in animal models and randomized clinical trials
mation is an important key to the pathophysiology of DKD. show efficacy of RAAS inhibition to slow the progression of
Nuclear factor kappa-light-chain-enhancer of activated B DKD.31 Both transforming growth factor-b1 (TGF-b1) and
cells (NF-kB), a transcription factor, is also a major regu- angiotensin II are involved in the processes of renal tissue
lator of inflammatory factors. In DKD, increased tumor fibrosis, including renal tubules dysfunction and atrophy,
necrosis factor (TNF) and ROS activate NF-kB by phos- reduction of the number of small vessels, and chronic
phorylation. Moreover, NF-kB is associated with proteinuria hypoxia.32 Aldosterone is also thought to play important
and renal interstitial inflammatory cells infiltration.21 roles in the pathophysiology of DKD, through regulating
The response of immune system is also related the gene expression and other mechanisms, including upregu-
development of DKD. The predominant immune response in lation of pro-sclerotic growth factors, such as plasminogen
the development of DKD is innate immune response.22 Hy- activator inhibitor 1 (PAI-1) and TGF-b, and promotion of
perglycemia causes cellular stress and dysfunction due to macrophage infiltration and consequent renal fibrosis.33
disturbed function of mitochondria and endoplasmic retic-
ulum, ROS production, and abnormal activation of intra-
cellular signals pathways. As a result of these diabetic Novel molecular pathogenesis
stress, kidney cells produce proinflammatory responses and
then facilitate innate immune response via release of Recent molecular and cellular researches explore novel
chemokines, cell adhesion molecules (CAMs) and danger fields of pathophysiology of diabetes and DKD, including
associated molecular patterns (DAMPs).23 The initiation of genetic and epigenetic regulation, mitochondria dysfunc-
innate immune response is recruitment of macrophage.24 tion and podocyte autophagy. By these mechanisms, the
As hyperglycemia persists, AGEs and oxidized lipoproteins targets of DKD treatment can be more specified (Fig. 3).
are increased and then cause formation of immune com-
plexes. These immune complexes deposit in the kidney and Genetic and epigenetic regulations
then further amplify the innate immune response by acti- Growing evidences support the important roles of genetic
vation of complement and immunoglobulin Fcg receptors. and epigenetic mechanisms of pathophysiology of DKD.
Activated complement facilitates the recruitment of leu- Epigenetic modifications consist of DNA methylation, non-
kocytes and mast cells in the kidney.25 As DKD continues coding RNAs and histone posttranslational modifications
developing, the susceptibility of renal ischemia becomes (PTMs).34 Hyperglycemia, hypoxia, inflammation, and cy-
more apparent, which enhances renal inflammation tokines are proved to be able to modify the epigenetic
through increasing infiltration of neutrophils and macro- profiles.35
phages in the kidney. These immune responses accelerate Several genes have different degrees of DNA methylation
renal damage and decline of renal function in DKD. Hence, between type 2 DM patients with DKD and without DKD. The
the inflammation in DKD is involved with a complex inte- gene UNC13B has been shown to involve the initial patho-
gration of innate immune responses.22 genesis of DKD and apoptosis of glomerular cells due to
Janus kinaseesignal transducer and activator of tran- hyperglycemia.36 Moreover, hypermethylation of RASALI
scription (JAKeSTAT) pathway has found to be integrally gene increases Ras activation in fibroblasts and then leads
associated with DKD progression. JAKeSTAT proteins can tissue proliferation and fibrosis.37 These studies prove the
assist innate immune responses in kidney cells through importance of DNA methylation in regulating genes related
chemokine, cytokine and angiotensin receptors. Animal to DKD, and provide possible therapeutic genetic targets of
diabetic model shows that selectively enhanced expression DKD management.
of JAK2 in glomerular podocytes increases functional and MicroRNAs (miRNAs) are small noncoding RNA, which
pathological features of DKD. Whether early or progressive promote translation repression or mRNA degradation.38
DKD, the expressions of JAKeSTAT genes are increased in Several miRNAs are considered to have effects on kidneys
glomerular podocytes and tubulointerstitium.26 because they are abundant in the kidneys. Among DKD-
Recent researches have focused on the roles of gut related miRNAs, miRNA-192 is the best studied one. The
microbiome, which is associated with chronic inflammation level of miRNA-192 increases in the early stage of DKD, and
status in type 2 DM and CKD. Gut dysbiosis, or pathogen miRNA-192 causes upregulation of the key genes associated
overgrowth, may promote loss of gut barrier integrity with the pathogenesis of DKD, including Cola2 and Col4a1,
(leaky gut syndrome), facilitate translocation of gastroin- in mesangial cells.39 Besides, miRNA-192 can promote the
testinal bacteria and its products into blood, and trigger the autoregulation of TGF-b1 in mesangial cells by regulating
activation of innate immune responses and a persistent other miRNAs and amplify the response of p53 to TGF-
systemic inflammation, which is possibly associated with b1.40,41
the development and progression of DKD.27 Histone PTMs regulate gene expression via lysine acet-
ylation, lysine methylation and phosphorylation. Histone
Overactive renin-angiotensin-aldosterone system acetylation is involved in the induction of miRNA-192 in
Hyperglycemia and AGEs induce expression of renin and DKD. The regulation of miRNAs is mediated changes in
angiotensinogen in renal cells via ROS and kidney specific G epigenetic histone acetylation under normal and diabetic
protein-coupled metabolic receptor GPR91.28,29 Besides, states.42 Histone methylation is associated with gene
high-sodium intake can blunt the antihypertensive and expression of TGF-b1-mediated extracellular matrix (ECM)
antiproteinuric effects of ACEi and ARB due to activation of expansion under normal and high glucose conditions.43
the renal renin-angiotensin system.30 Activation of intra- Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a
renal RAAS aggravates the deterioration of DKD. Evidences transcription factor of the basic region leucine zipper
666 Y.-C. Lin et al.

Figure 3 Novel molecular and cellular pathophysiology of diabetes kidney disease. Nrf2, nuclear factor (erythroid-derived 2)-
like 2; TGF-b1, transforming growth factor b1; PTMs, posttranslational modifications; Atg, autophagy-related protein; mTOR,
mammalian target of rapamycin; AMPK, adenosine monophosphate activated protein kinase; PI3K-I/PKB, phosphatidylinositol
3-kinase I/protein kinase B; eNOS, endothelial nitric oxide synthase; NF-kB, nuclear factor kappa-light-chain-enhancer of activated
B cells.

family.44 Nrf2 has important functions for regulating and of insulin or growth factors and interacts with insulin re-
activating the intracellular antioxidation to neutralize ROS, ceptors or tyrosine kinase receptors. Activation of Akt/PKB
and maintaining redox homeostasis. Several investigations and PKD1 (polycystic kidney disease 1), and subsequent
revealed protective effects of Nrf2 in diabetes through in- inhibition of tuberous sclerosis complex 1 and 2 (TSC1/2)
hibition of TGF- b1, decreased ECM production and down- promote mTOR activation. Finally, podocyte autophagy is
regulating cyclin-dependent kinase inhibitor 1A.45e48 suppressed.55
AMPK is an essential regulator in energy metabolism and
Podocyte autophagy can be activated by an increase in cellular calcium con-
Recent studies show that autophagy is involved in the centration and the stimulation of numerous hormones,
pathogenesis of diabetes-related podocyte injury and adipokines, and cytokines.56 In the condition of ATP defi-
renoprotective in diabetic nephropathy.49 Autophagy con- ciency, including starvation or insulin effect, the down-
trols the quality of the cytoplasm by degrading proteins, stream TSC1/2 is activated by AMPK, and finally enhances
peroxidases, and maintaining the homeostasis of intracel- autophagy by suppressing mTOR activation.57
lular environment.50 Activated podocyte autophagy has a
protective effect on DKD through autophagy-related (Atg) Mitochondria dysfunction
protein conjugation system and mammalian target of Traditional view on the association with mitochondria
rapamycin (mTOR) regulation. dysfunction and complications of DM involves hyperglyce-
Atg12 is a ubiquitin-like protein involving in the auto- mia-related deleterious effect. Hyperglycemia promotes
phagosome formation, and autophagy activation needs the overload of the electron transport chain and subsequently
conjugation of Atg12 to Atg5.51 b-arrestins is a negative causes DNA damage and reduced GAPDH activity via
adaptor of G protein-coupled receptors (GPCRs) and ag- increased ROS production. Hence, the course of glycolysis is
gravates podocyte injury through autophagy inhibition via disturbed and an accumulation of all upstream glycolytic
downregulating Atg12-Atg5 conjugation in DKD. Therefore, intermediates occurs. Metabolic flux is shifted into other
modulation of this pathway may be a novel therapeutic alternative processes, including increased activity in the
approach for treating patients with DKD.52 polyol pathway, production of AGE precursors, PKC acti-
mTOR is essential to regulation of cell growth, and vation, and hexosamine pathway activity. As a result,
activation of mTOR can suppress autophagy. In the up- fibrosis, thrombosis, DNA damage, cellular dysfunction,
stream of mTOR, there are two separated protein kinases, vascular leakage, angiogenesis and inflammation, and
phosphatidylinositol 3-kinase I (PI3K-I)/protein kinase decreased oxidative stress defense are accompanied to
B(Akt/PKB) and AMP-activated protein kinase (AMPK), cause kidney damages.58,59
which are regulated by different conditions.53 PI3Ks are Alternative mechanism of mitochondria involvement in
consisted of three isoforms, including PI3K-I, PI3K-II, and diabetic kidney is associated with reduced mitochondrial
PI3K-III.54 PI3K-I inhibits autophagy while PI3K-III activates superoxide. The view of high degree of ROS related to
it. The activation of class I PI3K is triggered by interaction harmful to many kinds of cells is well-known before. The
Update of pathophysiology and management of diabetic kidney disease 667

diabetic kidney was found to have a reduction of mito- for 9 years, microalbuminuria still occurred in 16% of pa-
chondrial content and reduced activity of the electron tients in the primary prevention group and 26% of patients
chain complexes. Normal mitochondrial superoxide level is in the secondary prevention group.66 Hence, other stra-
able to maintain AMPK activation and eNOS phosphoryla- tegies of treatment are still required to decrease the
tion. Hence, production of mitochondrial superoxide can be burden of DKD.
an indicator of healthy mitochondria and physiologic For patients with type 1 or type 2 DM, a number of risk
oxidative phosphorylation. In response to excess glucose factors related to deterioration from microalbuminuria to
exposure or nutrient stress, there is a reduction of mito- overt diabetes nephropathy consist of urinary albumin
chondrial superoxide, oxidative phosphorylation, and excretion, glycated hemoglobin (HbA1c) concentration,
mitochondrial ATP generation in the kidney. Decreased smoking, hypertension, and serum cholesterol concentra-
mitochondrial superoxides cause reduced AMPK activity and tions.67,68 Intensified multi-factorial interventions,
eNOS phosphorylation, and then subsequently stimulate including medical control of hyperglycemia, hypertension,
inflammatory processes via NF-kB and vascular dysfunc- dyslipidemia, and microalbuminuria, slow progression to
tion.60 Persistent reduction of mitochondrial oxidative overt diabetic nephropathy, retinopathy, autonomic neu-
phosphorylation complex activity is associated with the ropathy and ESRD in microalbuminuric patients with type 2
release of oxidants, proinflammatory and profibrotic cyto- DM.62,69
kines from nonmitochondrial sources. Finally, a manifes- A large prospective observational study indicated that
tation of organ dysfunction occurs.61 intensive glycemic control played an important role for
According to these theories, higher or lower mitochon- deterioration of diabetic nephropathy.70 The Action in
drial ROS levels, a biphasic response, increase the risks of Diabetes and Vascular Disease: Preterax and Diamicron
tissue damage. This concept is in the term of mitochondrial Modified Release Controlled Evaluation (ADVANCE) trial
hormesis.61 Stimulation of mitochondrial biogenesis and showed that intensive glucose control can reduce devel-
mitochondrial electron transport chain activity leads to opment of microalbuminuria and macroalbuminuria.62
beneficial outcomes in DKD. Restoration of mitochondrial In conclusion, glucose control in diabetes patients
function and superoxide production via activation of AMPK should be individualized according to the patient’s alert of
has now been associated with improvement in markers of hypoglycemia, underlying CKD or cardiovascular disease
renal dysfunction in diabetes. Approaches that stimulating status, and age. The National Kidney Foundation’s Kidney
AMPK via exercise, caloric restriction, or medicines can Disease Outcomes Quality Initiative (KDOQI) guideline
result in increasing mitochondrial oxidative phosphoryla- generally suggests keep HbA1c levels to 7.0% or less for
tion activity, restoring physiologic mitochondrial superox- patients with type 1 or type 2 diabetes mellitus. For in-
ide production, and promoting organ healing.60 dividuals with multiple co-morbidities, limited life expec-
tancy and risks of hypoglycemia episodes, target HbA1c
levels can be extended above 7.0%.71
Prevention and management

Interventions of DKD mainly consist of correction of hy- Blood pressure control


perglycemia, hypertension, and dyslipidemia, and modifi-
cation of lifestyle.62,63 Primary prevention represents Animal studies showed that controlling the blood pressure
prevention from normoalbuminuria to microalbuminuria, to normal level improved glomerular capillary hypertension
while secondary prevention represents prevention from in diabetic rats and can effectively reduce the incidence of
microalbuminuria to macroalbuminuria. Multiple interven- proteinuria and focal segmental glomerular lesions.72 Some
tional managements with control of blood glucose, blood observational studies suggested that the glomerular hy-
pressure and lipid, and smoking cessation can significantly pertension is the major cause of diabetic glomerulopathy.
improve the prognosis of cardiovascular events and help to Reducing systemic blood pressure without control glomer-
slow down the progression of renal disease, including ular capillary pressure simultaneously is insufficient to
macroalbuminuria and decreased eGFR for patients with prevent from glomerular damage.73
type 2 DM and DKD.64 Previous randomized trials revealed that angiotensin-
converting enzyme inhibitor (ACEi) is beneficial to reduce
Glucose control occurrence of microalbuminuria in normotensive and nor-
moalbuminuric patients with type 1 and type 2 DM. United
When normoalbuminuric patients with type 1 DM receive Kingdom Prospective Diabetes Study (UKPDS) showed that
glycemic control that achieving near-normal blood sugar microalbuminuria only occurred in a small portion of pa-
level, glomerular hyperfiltration may be corrected after tients with strict blood pressure control for 6 years follow-
short period. Meta-analysis showed that patients with type up. Besides, strict control of blood pressure can signifi-
1 DM receiving intensive blood glucose control for 8e60 cantly reduce incidence of microalbuminuria by 29%
months reduced the risk of progression of nephropathy.65 (p < 0.009).74
DCCT (Diabetes Control and Complications Trial) study RAAS inhibitors reduce the frequency of micro-
showed that intensified blood glucose control reduced albuminuria in hypertensive normoalbuminuric patients.
albuminuria, and EDIC (Epidemiology of Diabetes In- This effect is not significant for patients with normal blood
terventions and Complications) study indicated that pressure. It is likely that intrarenal RAAS is enhanced in
intensive DM therapy reduced the risk of an impaired GFR patients with hypertension, but not in those without hy-
by 50%. Even receiving intensified blood glucose control pertension. Hence, ACEi or other anti-hypertensive drugs
668 Y.-C. Lin et al.

are not suggested to use for primary prevention of DKD in morbidity and mortality in this population. Some statin
normoalbuminuric patients with normal blood pressure.75 seems to have benefits with improving proteinuria in pa-
RAAS blockers have the benefits for changes of glomer- tients with DKD. In the Prospective Evaluation of Protein-
ular structures in patients with early stage of DKD. ACEi and uria and Renal Function in Diabetic Patients With
angiotensin receptor blocker (ARB) can significantly lower Progressive Renal Disease trial (PLANET I trial), atorvastatin
blood pressure and urinary albumin excretion rate for has more renoprotective effects with reducing proteinuria
microalbuminuric patients with type 2 DM. Anti- and renal events than rosuvastatin in patients with DKD
hypertensive medications is beneficial to disease itself under ACEi or ARB treatment.88 KDOQI guideline recom-
and cost effectiveness for microalbuminuric patients with mends that patients with DKD should receive statin-based
DM.76e78 Long-term intensified multi-factorial interven- therapies to lower low-density lipoprotein cholesterol
tional treatment can reduce 50% of microvascular and (LDL-C) levels, to reduce risks of major atherosclerotic
cardiovascular events, decrease incidence of ESRD and events.71 Recent American College of Cardiology and the
absolute risks of death by 20% in microalbuminuric patients American Heart Association (ACC/AHA) blood cholesterol
with type 2 DM.79 guideline suggests that use the most tolerated statin dosage
RAAS blockers have effects of relieving occurrence of for primary and secondary prevention according to the
renal diseases for patients with type 1 or type 2 DM. ACEi degrees of patient’s atherosclerotic cardiovascular disease
and ARB are considered to have similar effects on lowering (ASCVD) risks. For all patients older than 21 years of age
blood pressure and decreasing albuminuria.80 Present rec- with primary LDL-C level more than 190 mg/dL and those
ommendations suggest that RAAS blockers can relieve the 40e75 years of age with DM and LDL-C level between 70 and
deterioration of nephropathy for albuminuric patients with 189 mg/dL, statin therapy should be initiated. Among pa-
DM, hypertension and advanced chronic kidney tients with DM who are younger than 40 years or older than
disease.63,81,82 75 years of age, or whose LDL-C level is less than 70 mg/dL,
Combination of ACEi and ARB has additional renal and statin therapy should be individualized by the ASCVD risk-
cardiovascular protective effects for albuminuric patients reduction benefits, the potential adverse effects, and pa-
with DM.83 Ongoing Telmisartan Alone and in Combination tient preferences.89
With Ramipril Global Endpoint Trial (ONTARGET) showed
that the combination of telmisartan and ramipril reduced
albuminuria to normal range but increased the risk of Life style modifications
doubling the concentration of serum creatinine, needs to
receive renal replacement therapy and mortality.84 Veter- For all diabetic patients, lifestyle modifications, including
ans Affair Nephro Diabetes (VA Nephron D) study also dietary restriction of sodium and protein, adequate exer-
revealed there is no significant difference between com- cise and weight reduction, and smoking cessation, are
bination of ACEi and ARB and single ACEi or ARB treatment. important to lower the risk of diabetic kidney disease and
Instead, combined therapy may increase the risks of cardiovascular events.
hyperkalemia and acute kidney injury.85 Individualized diet planning according to disease
Direct renin inhibitor, aliskiren, has been used to treat severity, blood pressure, electrolyte status and cultures are
hypertension, which can inhibit initial rate-determining suggested for patients with DKD in the effective educa-
step of RAAS. However, many large clinical trials indicate tional program because of high prevalence of multiple
that it is not recommended to use combined therapy of metabolic abnormalities.90 It is important that dietary so-
aliskiren and ACEi or ARB to patients with DM and hyper- dium restriction can give the most benefit in proteinuric
tension or renal insufficiency. The above combined therapy patients with DM.91 However, it still debated whether the
not only lacks significant benefit for prognosis, but also restrictive protein diet slows the progression of DKD. Ac-
increases the risks of hyperkalemia, hypotension, acute cording to the potential benefits and known risks of various
kidney injury and even cardiovascular events. Hence, Food protein diet, the Kidney Disease Improving Global Out-
and Drug Administration (FDA) and European Medicines comes (KDIGO) and the National Kidney Foundation (NKF)
Authority (EMA) regard the combined therapy with aliskiren suggest a target dietary protein intake of 0.8 g/kg/day for
plus ACEi or ARB as contraindication of patients with DM or people with or without diabetes and eGFR less than 30 ml/
renal insufficiency.86 min/1.73 m2.90
The recent Kidney Disease: Improving Global Outcome Physical activity has been considered to improve physical
(KDIGO) guideline for patients with DKD suggests a target health, blood pressure, lipid profiles, insulin sensitivity, and
blood pressure of 140/90 mmHg or less for all for all dia- cardiovascular outcomes in patients with diabetes.92,93 Be-
betic patients, and 130/80 mmHg or less for micro- sides, regular exercise has been shown to improve protein-
albuminuric patients with DM. Besides, use of ACEi or ARB is uria in rat models of diabetes.94 By the known overall health
recommended for patients with DKD and urine albumin benefits of physical activity, the American Diabetes Associ-
excretion of 30 mg per 24 h or more.87 ation suggests at least 150 min per week of moderate to
vigorous aerobic exercise at least 3 days a week for patient
with type 2 diabetes.95 Some small investigations have been
Lipid profiles control shown the benefit effect of weight loss on DKD by low-calorie
diet or bariatric surgery for obesity. For patients with dia-
Dyslipidemia is common in patients with DKD. Diabetic betes and obesity, weight reduction seems a positive strat-
patients with persistent albuminuria may increase the risks egy for DKD. It needs larger researches to understand the
of cardiovascular events, which are the common causes of association with DKD and obesity or weight loss.96,97
Update of pathophysiology and management of diabetic kidney disease 669

Smoking in particular is an independent risk factor for (hazard ratio Z 0.64; p Z 0.005).104 By these recent
the development of nephropathy in type 2 diabetes. Many studies results, GLP-1 analogs have benefits to DKD devel-
researches indicate the significantly negative effect of opment or progression.
smoking on cardiovascular events and global health. KDIGO Dipeptidyl peptidase 4 (DPP4) inhibitors linagliptin and
guideline encourage to avoid and quit smoking as possible saxagliptin (SAVOR-TIMI 53 trial) can reduce the amount of
for patients with established and at risk of DKD.87 albuminuria.105,106 However, whether DPP4 inhibitors have
more effectiveness on renal protection beyond decrease of
blood glucose level still needs further research to confirm
Novel treatments of diabetic kidney disease the clinical outcomes.
Some studies revealed that rosiglitazone, a kind of
Some novel treatments of DKD are developed and investi- thiazolidinedione, reduced microalbuminuria independent
gated in recent years (Table 2). on the hyperglycemic state.107,108

Glucose-lowering agents Other novel agents for DKD


Some glucose-lowering agents for patients with type 2 DM, Pentoxifylline (PTF) has been shown to attenuate the pro-
especially novel ones, are found to be able to protect gression of renal fibrosis through inhibition of cell prolif-
kidney other than the effect of controlling hyperglycemia. eration, reducing kidney inflammation and decreasing the
Renal sodium glucose cotransporter 2 (SGLT2) is accumulation of extracellular matrix in animal
responsible for reabsorption of the glucose in the glomer- studies.109,110 In recent studies enrolling 169 patients with
ular infiltrate, and its inhibitors have been used for type 2 type 2 DM and following up 2 years, it displayed that the use
DM. In patients with type 2 DM and high cardiovascular of RAAS plus PTF relieved the reduction of GFR and
risks, empagliflozin, an SGLT2 inhibitor, is associated with decreased urinary albumin excretion.111e113
slower progression of kidney disease and lower rates of Vitamin D or vitamin D analogs play important roles on
clinically relevant renal events than placebo when added to inhibition of occurrence of DKD. Vitamin D is a negative
standard care.74,98 Another SGLT2 inhibitor canagliflozin, in regulator of RAAS.114 In experimental studies, combined
a recently published trial (CANVAS trial), showed benefit of therapy with vitamin D analogs and ARB prevented from
canagliflozin with respect to the progression of albuminuria kidney injury and is more effective than any single one
(hazard ratio 0.73) and the composite outcome of a sus- use.115 In short-term study assessing safety of the use of
tained 40% reduction in the estimated GFR, the need for vitamin D analogs, paricalcitol had proteinuria-lowering
RRT, or death due to renal causes (hazard ratio 0.60).99 effect in patients with CKD.116 VITAL (Selective Vitamin D
According to these recent studies, SGLT2 inhibitors have Receptor Activator for Albuminuria Lowering) study
benefits for delaying DKD progression. enrolled patients with type 2 DM and albuminuria, and
Glucagon-like peptide-1 (GLP-1) is one of the incretins, showed that the addition of 2 mg paricalcitol to RAAS inhi-
released from the intestine in response to food intake. GLP- bition reduced albuminuria by 18%e28% (p Z 0.014 vs
1 can stimulate insulin secretion and then keep stable ho- placebo). However, the long-term effects are still
meostasis of glucose and GLP-1 level is decreased in pa- lacking.117
tients with type 2 diabetes. Several GLP-1 analogs have Pyridoxamine belongs to the family of vitamin B6. It can
been used to treat type 2 DM. In one study of 23 patients remove free radicals and carbonyl products, and block
with type 2 DM and overt DN, who had already been treated synthesis of AGEs. Pyridoxamine phase II trial showed that
with blockade of renin-angiotensin system under dietary although the serum creatinine level did not have difference
sodium restriction, GLP-1 analog liraglutide for a period of after using pyridoxamine for 52 weeks, but subgroup anal-
12 months caused a significant decrease in proteinuria ysis showed that patients with normal renal function had
(p Z 0.002) and substantially diminished the rate of decline lower average serum creatinine level after using pyridox-
in eGFR (p Z 0.003).100 In another study including 31 pa- amine. Currently, PIONEER-CSG-17 trial is trying to prove
tients with type 2 diabetes, short-term or long-term (one such benefits about use of pyridoxamine.118 On contrast, in
year) treatment of liraglutide were associated with signif- a randomized, double-blind placebo control trial of 238
icant reversible decline in GFR, and reduction of 27% urine participants with type 1 or type 2 diabetes and DN, patients
albumin excretion rate (p Z 0.020) and systolic blood taking high dose of B-vitamin containing folic acid (2.5 mg/
pressure (p Z 0.048).101 According to the LEADER (Lir- day), vitamin B6 (25 mg/day) and vitamin B12 (1 mg/day)
aglutide Effect and Action in Diabetes: Evaluation of Car- decreased more GFR significantly than those taking placebo
diovascular Outcomes Results) double-blind trial with 9340 (p Z 0.02). It needs larger and systemic investigations to
patients with type 2 DM and high cardiovascular risk, pa- confirm this view of point.119
tients with liraglutide had lower rate of the first occurrence Avosentan, an endothelin-1 receptor A antagonist, can
of death from cardiovascular causes, nonfatal myocardial reduce urinary albumin excretion. However, the associated
infarction, nonfatal stroke, and new onset of persistent study was terminated early because of excessive cardio-
macroalbuminuria (hazard ratio Z 0.74, p Z 0.004) than vascular events during the treatment course.120 Another
with those with placebo. In this trial, liraglutide also caused study of 211 patients showed atrasentan, a kind of selective
lower rates of progression of DKD than placebo.102,103 endothelin receptor A antagonist, had proteinuria-lowering
Another randomized study of 3297 patients with type 2 effect and less accumulation of body fluid.10
diabetes under standard-care regimen, patients using In a phase 2 randomized controlled trial for patients with
semaglutide (another GLP-1 analog) had lower rates of new type 2 DM, ruboxistaurin, an inhibitor of protein kinase C-b,
or deteriorating nephropathy than those with placebo significantly reduced albuminuria without decrease of
670 Y.-C. Lin et al.

Table 2 Novel medical treatments for diabetic kidney disease (DKD).


Medication & recent studies Action mechanism/molecular targets Study population/effects on renal outcomes
Glucose-lowering agents
SGLT2 inhibitors:
Empagliflozin Inhibition of SGLT-2 to decrease Type 2 DM and high CV risks
(EMPA-REG OUTCOME)98 glucose reabsorption Lower rates of incident or deteriorating
nephropathy, doubling of serum creatinine
level and RRT.
Canagliflozin (CANVAS)99 Inhibition of SGLT-2 to decrease Type 2 DM and high CV risks
glucose reabsorption Possible benefit with progression of
albuminuria, reduction of eGFR, need of
RRT and death from renal causes.
GLP-1 RAs:
Liraglutide (LEADER)103 Enhance GLP-1 expression to increase Type 2 DM and high CV risks
insulin secretion Lower rates of progression and development
of DKD
Semaglutide (SUSTAIN-6)104 Enhance GLP-1 expression to increase Type 2 DM and CV risks or CKD stage3
insulin secretion Lower rates of new or deteriorating
nephropathy
DPP-4 inhibitors:
Linagliptin105 Inhibition of DPP-4 to preserve GLP Type 2 DM with microalbuminuria or higher
effect and receiving stable dose of RAAS inhibitors
Reduction in albuminuria
Saxagliptin Inhibition of DPP-4 to preserve GLP Type 2 DM with albuminuria
(SAVOR-TIMI 53 trial)106 effect Improved albuminuria
Thiozolidinediones:
Rosiglitazone108 Activation of PPARg to increase Type 2 DM with albuminuria
insulin sensitivity of tissue Decreased albuminuria
Other novel agents
Phosphodiesterase inhibitors:
Pentoxifylline109,110,112 Inhibition of cell proliferation, kidney Type 2 DM and CKD stage 3e4 with RAAS
(PREDIAN trial) inflammation and accumulation of inhibitors
extracellular matrix Less decreased in eGFR; higher reduction of
albuminuria
Vitamin D analogs:
Paricalcitol116,117 Inhibition of RAAS Type 2 DM and albuminuria with RAAS
(VITAL study) inhibitors
Reduction of albuminuria
Pyridoxamine118 Remove free radicals and carbonyl Type 1 and type 2 DM with overt DN
products; block synthesis of AGEs Significant reduction of the changes of
serum creatinine
Endothelium A Type 2 DM and CKD stage 2e3
receptor antagonists:
Atrasentan10 Selective endothelin receptor A Reduced albuminuria
antagonist
Protein kinase C inhibitor:
Ruboxistaurin121 Inhibition of protein kinase C-b and Type 2 DM and albuminuria
reduce of oxidative stress Reduced albuminuria and maintain eGFR
Sulodexide123 Restores the anionic heparan sulfate Type 1 and type 2 DM with albuminuria
charges on the glomerular basement Reduced albuminuria
membrane
TGF-b blockade
Pirfenidone125 Antagonize MAPK pathway to Animal study
attenuate EMT and fibrosis Reduce the decline of GFR
Anti-inflammation
Bardoxolone methyl126 Activation of Nrf2 and inhibit NF-kB Type 2 DM and impaired renal function
pathway No influence of albuminuria
Update of pathophysiology and management of diabetic kidney disease 671

Table 2 (continued )
Medication & recent studies Action mechanism/molecular targets Study population/effects on renal outcomes
JAK inhibitor:
Baricitinib26 Selective JAK-1 and JAK-2 inhibitor to Type 2 DM and DKD
reduce innate immune response in Reduced albuminuria
kidney cells
Non-steroidal minerocorticoid antagonist:
Finerenone130 Inhibition of RAAS Type 2 DM and albuminuria with RAAS
inhibitors
Reduced albuminuria
SGLT2: Sodium/glucose cotransporter 2; GLP-1 RA: Glucagon-like peptide 1 receptor agonist; DPP-4: Dipeptidyl peptidase 4; PPARg:
peroxisome proliferators-activated receptor-gamma; MAPK: mitogen-activated protein kinases; EMT: epithelial to mesenchymal tran-
sition; Nrf2: nuclear factor (erythroid-derived 2) -like 2; JAK: Janus kinase; RRT: renal replacement therapy; eGFR: estimated
glomerular filtration rate.

estimated GFR.121 Nevertheless, the long-term renal out- had been conducted in type 2 diabetic participants with
comes and mortality rate of ruboxistaurin are similar to DKD. In this trial, there was a reduction of albuminuria in
that of placebo.122 receiving the active inhibitor. This result supports the
Sulodexide (Soludexide) is a kind of glycosaminoglycan further study of JAK inhibitors as a new therapy for DKD.26
(GAG) mixture of 20% of dermatan sulfate and 80% low- A study demonstrated that spironolactone, a miner-
molecular-weight heparin. The precise mechanism of the ocorticoid receptor antagonists (MRAs), in addition to ACE
renoprotective effects of sulodexide remains unknown. A inhibition, reduced proteinuria in patients with chronic
simple explanation of the efficacy of sulodexide is that it kidney disease (CKD) from either diabetes or nondiabetic
restores the anionic heparan sulfate charges on the glomer- causes.128 However, MRAs increase the risk of hyperkalemia
ular basement membrane. A recent meta-analysis showed in patients with stage 3 or more advanced chronic kidney
that sulodexide therapy was associated with a significant disease.129 Finerenone is a novel nonsteroidal MRA. In a
reduction in urinary protein excretion (Odds ratio is 3.28, randomized, double-blind, placebo-controlled study,
p Z 0.01) and had renoprotective benefit on patients with finerenone for 90 days showed a significant dose-dependent
diabetes and microalbuminuria and macroalbuminuria.123 reduction of albuminuria (p < 0.05) in the patients with
The development of other anti-fibrosis agents focuses on type 2 DM who had received ACEi or ARB.130 Other long-
antibodies against growth factors, including connective term or larger studies are needed to assure the efficacy
tissue growth factor (CTGF) and TGF-b blockade, for and safety of nonsteroidal MRA.
example, pirfenidone. Recent studies indicated that pirfe-
nidone had anti-fibrotic effects in many tissues. The po-
tential molecular mechanism remains unknown. Low dose Conclusion
pirfenidone reduces the decline of GFR. The associated
study was terminated due to more side effects at higher DKD is a clinical syndrome consisting of persistent albu-
dose of pirfenidone.124 On the other hand, in an animal minuria, sustained reduction in GFR, elevated blood pres-
study, pirfenidone was able to attenuate epithelial-to- sure, increased cardiovascular events and their related
mesenchymal transition (EMT) and fibrosis in vivo and mortality. The dialytic patients with DM have higher mor-
in vitro through antagonizing the mitogen-activated protein tality than those without DM. Recent studies showed that
kinases (MAPK) pathway, providing a potential treatment to multifactorial interventions with RAAS blockers, control of
alleviate renal tubulointerstitial fibrosis.125 blood pressure, blood glucose and lipid, and smoking
Laboratory studies showed that agents against inflam- cessation can significantly improve the prognosis of pa-
mation or oxidative stress may have potential effects on tients with type 2 DM and nephropathy. Many novel medi-
renal protection. Bardoxolone methyl reduces the genera- cations for prevention and treatment of DKD are studied,
tion of oxidative stress by the activation of nuclear factor including SGLT2 inhibitors, GLP-1 analogs, DPP-4 inhibitors,
(erythroid-derived 2) -like 2 (Nrf2) and inhibition of NF-kB thiazolidinedione, pentoxifylline, vitamin D analog par-
pathway. Short-term study revealed that the use of bar- icalcitol, pyridoxamine, ruboxistaurin, soludexide, JAK in-
doxolone methyl can increase the GFR in patients with type hibitors and nonsteroidal minerocorticoid receptor
2 DM and impaired renal function but have no influence of antagonists. In patients with DKD, further researches
albuminuria.126 However, the study was terminated early focusing on the renal outcomes, not only on changes of
because of more cardiovascular events.127 albuminuria, are needed to evaluate the long-term effects
According to the growing evidences of Janus of these novel medications.
kinaseesignal transducer and activator of transcription
(JAK-STAT) activation of DKD, baricitinib, a selective JAK-1
and JAK-2 inhibitor is developed. An unpublished Phase 2 Conflict of interest
multicenter, randomized, double-blind, placebo-controlled
study of the efficacy of a selective JAK1 and JAK2 inhibitor No conflict of interest to declared.
672 Y.-C. Lin et al.

Appendix A. Supplementary data 19. Forbes JM, Coughlan MT, Cooper ME. Oxidative stress as a
major culprit in kidney disease in diabetes. Diabetes 2008;
57(6):1446e54.
Supplementary data related to this article can be found at 20. Phisitkul K, Hegazy K, Chuahirun T, Hudson C, Simoni J,
https://doi.org/10.1016/j.jfma.2018.02.007. Rajab H, et al. Continued smoking exacerbates but cessation
ameliorates progression of early type 2 diabetic nephropathy.
Am J Med Sci 2008;335(4):284e91.
21. Ziyadeh FN, Wolf G. Pathogenesis of the podocytopathy and
References proteinuria in diabetic glomerulopathy. Curr Diabetes Rev
2008;4(1):39e45.
1. Gross JL, De Azevedo MJ, Silveiro SP, Canani LH, Caramori ML, 22. Wada J, Makino H. Innate immunity in diabetes and diabetic
Zelmanovitz T. Diabetic nephropathy: diagnosis, prevention, nephropathy. Nat Rev Nephrol 2016;12(1):13e26.
and treatment. Diabetes Care 2005;28(1):164e76. 23. Sourris KC, Forbes JM. Interactions between advanced gly-
2. de Boer IH, Rue TC, Cleary PA, Lachin JM, Molitch ME, cation end-products (AGE) and their receptors in the devel-
Steffes MW, et al. Long-term renal outcomes of patients with opment and progression of diabetic nephropathy - are these
type 1 diabetes mellitus and microalbuminuria: an analysis of receptors valid therapeutic targets. Curr Drug Targets 2009;
the diabetes control and complications trial/epidemiology of 10(1):42e50.
diabetes interventions and complications cohort. Arch Intern 24. Nguyen D, Ping F, Mu W, Hill P, Atkins RC, Chadban SJ.
Med 2011;171(5):412e20. Macrophage accumulation in human progressive diabetic ne-
3. Huang YY, Lin KD, Jiang YD, Chang CH, Chung CH, Chuang LM, phropathy. Nephrology (Carlton) 2006;11(3):226e31.
et al. Diabetes-related kidney, eye, and foot disease in 25. Flyvbjerg A. The role of the complement system in diabetic
Taiwan: an analysis of the nationwide data for 2000e2009. J nephropathy. Nat Rev Nephrol 2017;13(5):311e8.
Formos Med Assoc 2012;111(11):637e44. 26. Brosius FC, Tuttle KR, Kretzler M. JAK inhibition in the
4. 2014 Annual Report on Kidney Disease in Taiwan. http:// treatment of diabetic kidney disease. Diabetologia 2016;
www.tsn.org.tw/UI/H/2014ARKDT/2014_ARKDT_ch0.pdf. 59(8):1624e7.
5. Mogensen CE. How to protect the kidney in diabetic patients: 27. Anders HJ, Andersen K, Stecher B. The intestinal microbiota,
with special reference to IDDM. Diabetes 1997;46(Suppl 2): a leaky gut, and abnormal immunity in kidney disease. Kidney
S104e11. Int 2013;83(6):1010e6.
6. Retnakaran R, Cull CA, Thorne KI, Adler AI, Holman RR. Risk 28. Giacchetti G, Sechi LA, Rilli S, Carey RM. The renin-angio-
factors for renal dysfunction in type 2 diabetes: U.K. Pro- tensin-aldosterone system, glucose metabolism and diabetes.
spective Diabetes Study 74. Diabetes 2006;55(6):1832e9. Trends Endocrinol Metab 2005;16(3):120e6.
7. Melmed S, Polonsky KS, Larsen PR, Kronenberg HM. Williams 29. Toma I, Kang JJ, Sipos A, Vargas S, Bansal E, Hanner F, et al.
textbook of endocrinology. Elsevier Health Sciences; 2015. Succinate receptor GPR91 provides a direct link between high
8. Tuttle KR. Back to the future: glomerular hyperfiltration and glucose levels and renin release in murine and rabbit kidney. J
the diabetic kidney. Diabetes 2017;66(1):14e6. Clin Invest 2008;118(7):2526e34.
9. Potenza MA, Gagliardi S, Nacci C, Carratu MR, Montagnani M. 30. Vegter S, Perna A, Postma MJ, Navis G, Remuzzi G,
Endothelial dysfunction in diabetes: from mechanisms to Ruggenenti P. Sodium intake, ACE inhibition, and progression
therapeutic targets. Curr Med Chem 2009;16(1):94e112. to ESRD. J Am Soc Nephrol 2012;23(1):165e73.
10. de Zeeuw D, Coll B, Andress D, Brennan JJ, Tang H, Houser M, 31. Gurley SB, Coffman TM. The renin-angiotensin system and
et al. The endothelin antagonist atrasentan lowers residual diabetic nephropathy. Semin Nephrol 2007;27(2):144e52.
albuminuria in patients with type 2 diabetic nephropathy. J 32. He J, Xu Y, Koya D, Kanasaki K. Role of the endothelial-to-
Am Soc Nephrol 2014;25(5):1083e93. mesenchymal transition in renal fibrosis of chronic kidney
11. Prabhakar SS. Role of nitric oxide in diabetic nephropathy. disease. Clin Exp Nephrol 2013;17(4):488e97.
Semin Nephrol 2004;24(4):333e44. 33. Ritz E, Tomaschitz A. Aldosterone, a vasculotoxic
12. Bernhardt W, Schmitt R, Rosenberger C, Münchenhagen P, agentenovel functions for an old hormone. Nephrol Dial
Gröne H, Frei U, et al. Expression of hypoxia-inducible tran- Transplant 2009;24(8):2302e5.
scription factors in developing human and rat kidneys. Kidney 34. Portela A, Esteller M. Epigenetic modifications and human
Int 2006;69(1):114e22. disease. Nat Biotechnol 2010;28(10):1057e68.
13. Resnick HE, Redline S, Shahar E, Gilpin A, Newman A, Walter R, 35. Horsburgh S, Robson-Ansley P, Adams R, Smith C. Exercise and
et al. Diabetes and sleep disturbances: findings from the Sleep inflammation-related epigenetic modifications: focus on DNA
Heart Health Study. Diabetes Care 2003;26(3):702e9. methylation. Exerc Immunol Rev 2015;21:26e41.
14. Aronsohn RS, Whitmore H, Van Cauter E, Tasali E. Impact of 36. Bell CG, Teschendorff AE, Rakyan VK, Maxwell AP, Beck S,
untreated obstructive sleep apnea on glucose control in type Savage DA. Genome-wide DNA methylation analysis for dia-
2 diabetes. Am J Respir Crit Care Med 2010;181(5):507e13. betic nephropathy in type 1 diabetes mellitus. BMC Med Ge-
15. Peppard PE, Young T, Palta M, Skatrud J. Prospective study of nomics 2010;3:33.
the association between sleep-disordered breathing and hy- 37. Bechtel W, McGoohan S, Zeisberg EM, Muller GA, Kalbacher H,
pertension. N Engl J Med 2000;342(19):1378e84. Salant DJ, et al. Methylation determines fibroblast activation
16. Leong WB, Jadhakhan F, Taheri S, Thomas GN, Adab P. The and fibrogenesis in the kidney. Nat Med 2010;16(5):544e50.
association between obstructive sleep apnea on diabetic 38. Bartel DP. MicroRNAs: target recognition and regulatory
kidney disease: a systematic review and meta-analysis. Sleep functions. Cell 2009;136(2):215e33.
2016;39(2):301e8. 39. Kato M, Zhang J, Wang M, Lanting L, Yuan H, Rossi JJ, et al.
17. Pallayova M, Banerjee D, Taheri S. Novel insights into meta- MicroRNA-192 in diabetic kidney glomeruli and its function in
bolic sequelae of obstructive sleep apnoea: a link between TGF-beta-induced collagen expression via inhibition of E-box
hypoxic stress and chronic diabetes complications. Diabetes repressors. Proc Natl Acad Sci U S A 2007;104(9):3432e7.
Res Clin Pract 2014;104(2):197e205. 40. Kato M, Arce L, Wang M, Putta S, Lanting L, Natarajan R. A
18. Tan KC, Chow WS, Lam JC, Lam B, Bucala R, Betteridge J, microRNA circuit mediates transforming growth factor-beta1
et al. Advanced glycation endproducts in nondiabetic patients autoregulation in renal glomerular mesangial cells. Kidney Int
with obstructive sleep apnea. Sleep 2006;29(3):329e33. 2011;80(4):358e68.
Update of pathophysiology and management of diabetic kidney disease 673

41. Deshpande SD, Putta S, Wang M, Lai JY, Bitzer M, Nelson RG, 62. Perkovic V, Heerspink HL, Chalmers J, Woodward M, Jun M, Li Q,
et al. Transforming growth factor-beta-induced cross talk et al. Intensive glucose control improves kidney outcomes in
between p53 and a microRNA in the pathogenesis of diabetic patients with type 2 diabetes. Kidney Int 2013;83(3):517e23.
nephropathy. Diabetes 2013;62(9):3151e62. 63. Standards of medical care in diabetese2014. Diabetes Care
42. Kato M, Dang V, Wang M, Park JT, Deshpande S, Kadam S, 2014;37(Suppl 1):S14e80.
et al. TGF-beta induces acetylation of chromatin and of Ets-1 64. Andresdottir G, Jensen ML, Carstensen B, Parving HH,
to alleviate repression of miR-192 in diabetic nephropathy. Sci Rossing K, Hansen TW, et al. Improved survival and renal
Signal 2013;6(278):ra43. prognosis of patients with type 2 diabetes and nephropathy
43. Sun G, Reddy MA, Yuan H, Lanting L, Kato M, Natarajan R. with improved control of risk factors. Diabetes Care 2014;
Epigenetic histone methylation modulates fibrotic gene 37(6):1660e7.
expression. J Am Soc Nephrol 2010;21(12):2069e80. 65. Wang P, Lau J, Chalmers T. Meta-analysis of effects of
44. Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network intensive blood-glucose control on late complications of type I
provides an interface between redox and intermediary diabetes. Lancet 1993;341(8856):1306e9.
metabolism. Trends Biochem Sci 2014;39(4):199e218. 66. Group DER. Intensive diabetes therapy and glomerular filtra-
45. Yoh K, Hirayama A, Ishizaki K, Yamada A, Takeuchi M, tion rate in type 1 diabetes. N Engl J Med 2011;365(25):2366.
Yamagishi S, et al. Hyperglycemia induces oxidative and 67. Ahmad J, Siddiqui MA, Ahmad H. Effective postponement of
nitrosative stress and increases renal functional impairment diabetic nephropathy with enalapril in normotensive type 2
in Nrf2-deficient mice. Genes Cells 2008;13(11):1159e70. diabetic patients with microalbuminuria. Diabetes Care 1997;
46. Jiang T, Huang Z, Lin Y, Zhang Z, Fang D, Zhang DD. The 20(10):1576e81.
protective role of Nrf2 in streptozotocin-induced diabetic 68. Huang CY, Ting WH, Lo FS, Tsai JD, Sun FJ, Chan CI, et al.
nephropathy. Diabetes 2010;59(4):850e60. Factors associated with diabetic nephropathy in children,
47. Pergola PE, Krauth M, Huff JW, Ferguson DA, Ruiz S, Meyer CJ, adolescents, and adults with type 1 diabetes. J Formos Med
et al. Effect of bardoxolone methyl on kidney function in Assoc 2017;116(12):924e32.
patients with T2D and Stage 3b-4 CKD. Am J Nephrol 2011; 69. Gaede P, Vedel P, Larsen N, Jensen GV, Parving HH,
33(5):469e76. Pedersen O. Multifactorial intervention and cardiovascular
48. Xu X, Luo P, Wang Y, Cui Y, Miao L. Nuclear factor (erythroid- disease in patients with type 2 diabetes. N Engl J Med 2003;
derived 2)-like 2 (NFE2L2) is a novel therapeutic target for 348(5):383e93.
diabetic complications. J Int Med Res 2013;41(1):13e9. 70. Breyer JA, Bain RP, Evans JK, Nahman Jr NS, Lewis EJ,
49. Kim WY, Nam SA, Song HC, Ko JS, Park SH, Kim HL, et al. The Cooper M, et al. Predictors of the progression of renal insuf-
role of autophagy in unilateral ureteral obstruction rat model. ficiency in patients with insulin-dependent diabetes and overt
Nephrology (Carlton) 2012;17(2):148e59. diabetic nephropathy. The Collaborative Study Group. Kidney
50. Zhu J, Wang KZ, Chu CT. After the banquet: mitochondrial Int 1996;50(5):1651e8.
biogenesis, mitophagy, and cell survival. Autophagy 2013; 71. KDOQI Clinical practice guideline for diabetes and CKD: 2012
9(11):1663e76. update. Am J Kidney Dis 2012;60(5):850e86.
51. Geng J, Klionsky DJ. The Atg8 and Atg12 ubiquitin-like 72. Zatz R, Dunn BR, Meyer TW, Anderson S, Rennke HG,
conjugation systems in macroautophagy. ’Protein modifica- Brenner BM. Prevention of diabetic glomerulopathy by phar-
tions: beyond the usual suspects’ review series. EMBO Rep macological amelioration of glomerular capillary hyperten-
2008;9(9):859e64. sion. J Clin Invest 1986;77(6):1925e30.
52. Liu J, Li QX, Wang XJ, Zhang C, Duan YQ, Wang ZY, et al. beta- 73. Fujihara CK, Padilha RM, Zatz R. Glomerular abnormalities in
Arrestins promote podocyte injury by inhibition of autophagy long-term experimental diabetes. Role of hemodynamic and
in diabetic nephropathy. Cell Death Dis 2016;7. e2183. nonhemodynamic factors and effects of antihypertensive
53. Laplante M, Sabatini DM. mTOR signaling in growth control therapy. Diabetes 1992;41(3):286e93.
and disease. Cell 2012;149(2):274e93. 74. Efficacy of atenolol and captopril in reducing risk of macro-
54. Engelman JA, Luo J, Cantley LC. The evolution of phospha- vascular and microvascular complications in type 2 diabetes:
tidylinositol 3-kinases as regulators of growth and meta- UKPDS 39. UK Prospective Diabetes Study Group. BMJ 1998;
bolism. Nat Rev Genet 2006;7(8):606e19. 317(7160):713e20.
55. Sarbassov DD, Ali SM, Sabatini DM. Growing roles for the mTOR 75. Ali MK, Bullard KM, Saaddine JB, Cowie CC, Imperatore G,
pathway. Curr Opin Cell Biol 2005;17(6):596e603. Gregg EW. Achievement of goals in US diabetes care,
56. Zhang BB, Zhou G, Li C. AMPK: an emerging drug target for 1999e2010. N Engl J Med 2013;368(17):1613e24.
diabetes and the metabolic syndrome. Cell Metab 2009;9(5): 76. Palmer AJ, Annemans L, Roze S, Lamotte M, Lapuerta P,
407e16. Chen R, et al. Cost-effectiveness of early irbesartan treat-
57. Jin Y, Liu S, Ma Q, Xiao D, Chen L. Berberine enhances the ment versus control (standard antihypertensive medications
AMPK activation and autophagy and mitigates high glucose- excluding ACE inhibitors, other angiotensin-2 receptor an-
induced apoptosis of mouse podocytes. Eur J Pharmacol 2017; tagonists, and dihydropyridine calcium channel blockers) or
794:106e14. late irbesartan treatment in patients with type 2 diabetes,
58. Giacco F, Brownlee M. Oxidative stress and diabetic compli- hypertension, and renal disease. Diabetes Care 2004;27(8):
cations. Circ Res 2010;107(9):1058e70. 1897e903.
59. Du X, Matsumura T, Edelstein D, Rossetti L, Zsengeller Z, 77. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE,
Szabo C, et al. Inhibition of GAPDH activity by poly(ADP- Parving HH, et al. Effects of losartan on renal and cardio-
ribose) polymerase activates three major pathways of hy- vascular outcomes in patients with type 2 diabetes and ne-
perglycemic damage in endothelial cells. J Clin Invest 2003; phropathy. N Engl J Med 2001;345(12):861e9.
112(7):1049e57. 78. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB,
60. Sharma K. Mitochondrial hormesis and diabetic complications. et al. Renoprotective effect of the angiotensin-receptor
Diabetes 2015;64(3):663e72. antagonist irbesartan in patients with nephropathy due to
61. Gordon JW, Dolinsky VW, Mughal W, Gordon GR, McGavock J. type 2 diabetes. N Engl J Med 2001;345(12):851e60.
Targeting skeletal muscle mitochondria to prevent type 2 79. Mathiesen ER, Feldt-Rasmussen B, Hommel E, Deckert T,
diabetes in youth. Biochem Cell Biol 2015;93(5):452e65. Parving H-H. Stable glomerular filtration rate in normotensive
674 Y.-C. Lin et al.

IDDM patients with stable microalbuminuria: a 5-year pro- 95. Zinman B, Ruderman N, Campaigne BN, Devlin JT,
spective study. Diabetes Care 1997;20(3):286e9. Schneider SH. Physical activity/exercise and diabetes melli-
80. Lacourcière Y, Bélanger A, Godin C, Hallé J-P, Ross S, tus. Diabetes Care 2003;26(Suppl 1):S73e7.
Wright N, et al. Long-term comparison of losartan and ena- 96. Friedman AN, Chambers M, Kamendulis LM, Temmerman J.
lapril on kidney function in hypertensive type 2 diabetics with Short-term changes after a weight reduction intervention in
early nephropathy. Kidney Int 2000;58(2):762e9. advanced diabetic nephropathy. Clin J Am Soc Nephrol 2013;
81. Chen PM, Lai TS, Chen PY, Lai CF, Yang SY, Wu V, et al. 8(11):1892e8.
Multidisciplinary care program for advanced chronic kidney 97. Heneghan HM, Cetin D, Navaneethan SD, Orzech N,
disease: reduces renal replacement and medical costs. Am J Brethauer SA, Schauer PR. Effects of bariatric surgery on
Med 2015;128(1):68e76. diabetic nephropathy after 5 years of follow-up. Surg Obesity
82. Chen CH, Wu HY, Wang CL, Yang FJ, Wu PC, Hung SC, et al. Relat Dis 2013;9(1):7e14.
Proteinuria as a therapeutic target in advanced chronic kid- 98. Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M,
ney disease: a retrospective multicenter cohort study. Sci Rep Mattheus M, et al. Empagliflozin and progression of kidney
2016;6:26539. disease in type 2 diabetes. N Engl J Med 2016;375(4):323e34.
83. Jacobsen P, Andersen S, Rossing K, Jensen BR, Parving H-H. 99. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G,
Dual blockade of the renin-angiotensin system versus maximal Erondu N, et al. Canagliflozin and cardiovascular and renal
recommended dose of ACE inhibition in diabetic nephropathy. events in type 2 diabetes. N Engl J Med 2017;377(7):644e57.
Kidney Int 2003;63(5):1874e80. 100. Imamura S, Hirai K, Hirai A. The glucagon-like peptide-1 re-
84. Mann JF, Schmieder RE, McQueen M, Dyal L, Schumacher H, ceptor agonist, liraglutide, attenuates the progression of
Pogue J, et al. Renal outcomes with telmisartan, ramipril, or overt diabetic nephropathy in type 2 diabetic patients.
both, in people at high vascular risk (the ONTARGET study): a Tohoku J Exp Med 2013;231(1):57e61.
multicentre, randomised, double-blind, controlled trial. 101. von Scholten BJ, Hansen TW, Goetze JP, Persson F, Rossing P.
Lancet 2008;372(9638):547e53. Glucagon-like peptide 1 receptor agonist (GLP-1 RA): long-
85. Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, term effect on kidney function in patients with type 2 dia-
Duckworth W, et al. Combined angiotensin inhibition for the betes. J Diabetes Complications 2015;29(5):670e4.
treatment of diabetic nephropathy. N Engl J Med 2013; 102. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P,
369(20):1892e903. Mann JF, Nauck MA, et al. Liraglutide and cardiovascular
86. Sen S, Ufuktepe B, Ozunal ZG, Uresin Y. Renin inhibitors in outcomes in type 2 diabetes. N Engl J Med 2016;375(4):
diabetes and hypertension: an update. EXCLI J 2014;13: 311e22.
1111e9. 103. Mann JFE, Orsted DD, Brown-Frandsen K, Marso SP,
87. Taler SJ, Agarwal R, Bakris GL, Flynn JT, Nilsson PM, Poulter NR, Rasmussen S, et al. Liraglutide and renal out-
Rahman M, et al. KDOQI US commentary on the 2012 KDIGO comes in type 2 diabetes. N Engl J Med 2017;377(9):839e48.
clinical practice guideline for management of blood pressure 104. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E,
in CKD. Am J Kidney Dis 2013;62(2):201e13. Leiter LA, et al. Semaglutide and cardiovascular outcomes in
88. de Zeeuw D, Anzalone DA, Cain VA, Cressman MD, patients with type 2 diabetes. N Engl J Med 2016;375(19):
Heerspink HJ, Molitoris BA, et al. Renal effects of atorvastatin 1834e44.
and rosuvastatin in patients with diabetes who have pro- 105. Groop PH, Cooper ME, Perkovic V, Emser A, Woerle HJ, von
gressive renal disease (PLANET I): a randomised clinical trial. Eynatten M. Linagliptin lowers albuminuria on top of recom-
Lancet Diabetes Endocrinol 2015;3(3):181e90. mended standard treatment in patients with type 2 diabetes
89. Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, and renal dysfunction. Diabetes Care 2013;36(11):3460e8.
Blum CB, Eckel RH, et al. 2013 ACC/AHA guideline on the 106. Mosenzon O, Leibowitz G, Bhatt DL, Cahn A, Hirshberg B,
treatment of blood cholesterol to reduce atherosclerotic Wei C, et al. Effect of saxagliptin on renal outcomes in the
cardiovascular risk in adults: a report of the American College SAVOR-TIMI 53 trial. Diabetes Care 2017;40(1):69e76.
of Cardiology/American Heart Association Task Force on 107. Bakris GL, Ruilope LM, McMorn SO, Weston WM, Heise MA,
Practice Guidelines. J Am Coll Cardiol 2014;63(25 Pt B): Freed MI, et al. Rosiglitazone reduces microalbuminuria and
2889e934. blood pressure independently of glycemia in type 2 diabetes
90. Andrassy KM. Comments on ’KDIGO 2012 clinical practice patients with microalbuminuria. J Hypertens 2006;24(10):
guideline for the evaluation and management of chronic 2047e55.
kidney disease’. Kidney Int 2013;84(3):622e3. 108. Bakris G, Viberti G, Weston WM, Heise M, Porter LE, Freed MI.
91. Esnault VL, Ekhlas A, Delcroix C, Moutel MG, Nguyen JM. Rosiglitazone reduces urinary albumin excretion in type II
Diuretic and enhanced sodium restriction results in improved diabetes. J Hum Hypertens 2003;17(1):7e12.
antiproteinuric response to RAS blocking agents. J Am Soc 109. Lin SL, Chen RH, Chen YM, Chiang WC, Lai CF, Wu KD, et al.
Nephrol 2005;16(2):474e81. Pentoxifylline attenuates tubulointerstitial fibrosis by block-
92. Tanasescu M, Leitzmann MF, Rimm EB, Hu FB. Physical activity ing Smad3/4-activated transcription and profibrogenic effects
in relation to cardiovascular disease and total mortality of connective tissue growth factor. J Am Soc Nephrol 2005;
among men with type 2 diabetes. Circulation 2003;107(19): 16(9):2702e13.
2435e9. 110. Lin SL, Chen YM, Chiang WC, Tsai TJ, Chen WY. Pentoxifylline:
93. Salem MA, AboElAsrar MA, Elbarbary NS, ElHilaly RA, a potential therapy for chronic kidney disease. Nephrology
Refaat YM. Is exercise a therapeutic tool for improvement of (Carlton) 2004;9(4):198e204.
cardiovascular risk factors in adolescents with type 1 diabetes 111. Chen PM, Lai TS, Chen PY, Lai CF, Wu V, Chiang WC, et al.
mellitus? A randomised controlled trial. Diabetol Metab Syndr Renoprotective effect of combining pentoxifylline with
2010;2(1):47. angiotensin-converting enzyme inhibitor or angiotensin II re-
94. Tufescu A, Kanazawa M, Ishida A, Lu H, Sasaki Y, Ootaka T, ceptor blocker in advanced chronic kidney disease. J Formos
et al. Combination of exercise and losartan enhances reno- Med Assoc 2014;113(4):219e26.
protective and peripheral effects in spontaneously type 2 112. Navarro-Gonzalez JF, Mora-Fernandez C, Muros de Fuentes M,
diabetes mellitus rats with nephropathy. J Hypertens 2008; Chahin J, Mendez ML, Gallego E, et al. Effect of pentoxifylline
26(2):312e21. on renal function and urinary albumin excretion in patients
Update of pathophysiology and management of diabetic kidney disease 675

with diabetic kidney disease: the PREDIAN trial. J Am Soc 122. Tuttle KR, McGill JB, Haney DJ, Lin TE, Anderson PW. Kidney
Nephrol 2015;26(1):220e9. outcomes in long-term studies of ruboxistaurin for diabetic
113. Lai TS, Chiang WC, Chen YM. Pentoxifylline: evidence strong eye disease. Clin J Am Soc Nephrol 2007;2(4):631e6.
enough for renoprotection? J Formos Med Assoc 2016;115(8): 123. Li R, Xing J, Mu X, Wang H, Zhang L, Zhao Y, et al. Sulodexide
591e2. therapy for the treatment of diabetic nephropathy, a meta-
114. Li YC, Kong J, Wei M, Chen ZF, Liu SQ, Cao LP. 1,25-Dihy- analysis and literature review. Drug Des Devel Ther 2015;9:
droxyvitamin D(3) is a negative endocrine regulator of the 6275e83.
renin-angiotensin system. J Clin Invest 2002;110(2):229e38. 124. Sharma K, Ix JH, Mathew AV, Cho M, Pflueger A, Dunn SR,
115. Zhang Z, Zhang Y, Ning G, Deb DK, Kong J, Li YC. Combination et al. Pirfenidone for diabetic nephropathy. J Am Soc Nephrol
therapy with AT1 blocker and vitamin D analog markedly ame- 2011;22(6):1144e51.
liorates diabetic nephropathy: blockade of compensatory renin 125. Li Z, Liu X, Wang B, Nie Y, Wen J, Wang Q, et al. Pirfenidone
increase. Proc Natl Acad Sci U S A 2008;105(41):15896e901. suppresses MAPK signaling pathway to reverse epithelial-
116. Agarwal R, Acharya M, Tian J, Hippensteel RL, Melnick JZ, mesenchymal transition and renal fibrosis. Nephrology (Carl-
Qiu P, et al. Antiproteinuric effect of oral paricalcitol in ton) 2017;22(8):589e97.
chronic kidney disease. Kidney Int 2005;68(6):2823e8. 126. Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW,
117. de Zeeuw D, Agarwal R, Amdahl M, Audhya P, Coyne D, Grossman EB, et al. Bardoxolone methyl and kidney function
Garimella T, et al. Selective vitamin D receptor activation in CKD with type 2 diabetes. N Engl J Med 2011;365(4):
with paricalcitol for reduction of albuminuria in patients with 327e36.
type 2 diabetes (VITAL study): a randomised controlled trial. 127. de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-
Lancet 2010;376(9752):1543e51. Schmidt H, et al. Bardoxolone methyl in type 2 diabetes and
118. Williams ME, Bolton WK, Khalifah RG, Degenhardt TP, stage 4 chronic kidney disease. N Engl J Med 2013;369(26):
Schotzinger RJ, McGill JB. Effects of pyridoxamine in combined 2492e503.
phase 2 studies of patients with type 1 and type 2 diabetes and 128. Chrysostomou A, Becker G. Spironolactone in addition to ACE
overt nephropathy. Am J Nephrol 2007;27(6):605e14. inhibition to reduce proteinuria in patients with chronic renal
119. House AA, Eliasziw M, Cattran DC, Churchill DN, Oliver MJ, disease. N Engl J Med 2001;345(12):925e6.
Fine A, et al. Effect of B-vitamin therapy on progression of 129. Lazich I, Bakris GL. Prediction and management of hyper-
diabetic nephropathy: a randomized controlled trial. JAMA kalemia across the spectrum of chronic kidney disease. Semin
2010;303(16):1603e9. Nephrol 2014;34(3):333e9.
120. Mann JF, Green D, Jamerson K, Ruilope LM, Kuranoff SJ, 130. Bakris GL, Agarwal R, Chan JC, Cooper ME, Gansevoort RT,
Littke T, et al. Avosentan for overt diabetic nephropathy. J Haller H, et al. Effect of finerenone on albuminuria in patients
Am Soc Nephrol 2010;21(3):527e35. with diabetic nephropathy: a randomized clinical trial. JAMA
121. Tuttle KR, Bakris GL, Toto RD, McGill JB, Hu K, Anderson PW. 2015;314(9):884e94.
The effect of ruboxistaurin on nephropathy in type 2 dia-
betes. Diabetes Care 2005;28(11):2686e90.

You might also like