Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Clinical Queries: Nephrology 0102 (2012) 121–126

Contents lists available at ScienceDirect

Clinical Queries: Nephrology


j o u r n a l h o m e p a g e : h t t p : / / w w w. e l s e v i e r. c o m / l o c a t e / c q n

Pathophysiology of diabetic nephropathy


P.B. Vinod*
Consultant Nephrologist, Elite Mission Hospital, Thrissur, Kerala, India.

A R T I C L E I N F O A B S T R A C T

Article history: Diabetes is the major cause of chronic kidney disease which in turn may lead to end-stage renal disease
Received 11 February 2012 (ESRD) ending up in dialysis. Hemodynamic and structural changes following diabetes are working together
Accepted 1 March 2012 in the process of development of diabetic nephropathy (DN). Hyperglycemia-induced metabolic and hemo-
dynamic pathways are proven to be the mediators of kidney disease. Hyperglycemia causes the formation of
Keywords: Amadori products, which are the altered proteins and advanced glycation end products (AGE) are the molecu-
Diabetic nephropathy lar players in the phases of DN. According to recent studies, activation of electron transport chain induced
Glomerulosclerosis
by hyperglycemia can result in an increase in the reactive oxygen species (ROS) formation, which is thought
Pathophysiology
Tubulointerstitial fibrosis
to be the initiating event in the development of complications in diabetes. Hemodynamic changes, hyper-
trophy, extracellular matrix accumulation, growth factor/cytokine induction, ROS formation, podocyte dam-
age, proteinuria, and interstitial inflammation are the steps in the advancement of DN. High glucose, AGEs,
and ROS act in unison to induce growth factors and cytokines through signal transduction pathways involving
protein kinase C, mitogen-activated protein kinases, and the transcription factor NF-κB. Transforming growth
factor (TGF)-β causes hypertrophy of the renal cells and accumulation of extracellular matrix. Activation of
rennin angiotensin system with the subsequent formation of angiotensin-II (ANG-II) is involved in almost
all the steps in the development of DN. Blocking the action of ANG-II has a crucial role in every therapeutic
regimen to prevent and treat DN.
Copyright © 2012, Reed Elsevier India Pvt. Ltd. All rights reserved.

Introduction approaches.9,11 The recent studies suggest that the insertion or


deletion polymorphism of angiotensin-converting enzyme (ACE) is
The common cause for end-stage renal disease (ESRD) in developing associated only with the progression of the disease and it cannot be
countries is diabetic nephropathy (DN) and studies suggest that equated as predictor of the development of DN.8 A recent genome
there are chances of increase in this number in future.1,2 In contrast scan for DN in African Americans identified susceptibility loci on
to the previous belief that type 2 diabetes mellitus (DM) is a benign chromosomes 3q, 7p, and 18q.12 Another scan in Pima Indians also
condition related to normal ageing process, this is projected as the identified linkage to DN on chromosome 7.13 This powerful method
main single cause of renal failure in the USA, Japan, and Europe.3–5 may in the future more clearly identify the genetic risk to develop DN.
The present data showing 10 million people with diabetes will dou-
ble by 2030 in the US.6 The financial requirements for maintenance
of dialysis in patients with ESRD due to type 2 DM are higher com- Pathophysiology of diabetic nephropathy
pared to non-diabetic ESRD patients. The mortality of these patients
is higher owing to the cardiovascular complications.4,5 The thorough The pathophysiological mechanisms in the development of DN are
knowledge of the pathophysiology of this disease is very essential multifactorial. Hyperglycemia is the initiating event which causes
in order to develop newer therapeutic concepts for the prevention of structural and functional changes such as glomerular hyperfiltration,
DN.7 The renal injury in DN is due to a complex series of pathophysi- glomerular and tubular epithelial hypertrophy, and microalbuminu-
ological changes initiated by disturbed glucose homeostasis. ria, followed by the development of glomerular basement membrane
The basic pathophysiology of DN is similar in both type 1 and type 2 thickening, accumulation of mesangial matrix and overt proteinuria,
DM patients.2 Moreover, co-morbidities like hypertension, obesity, and finally glomerulosclerosis and ESRD.
dyslipidemia, and arterial sclerosis impart more injury to the kidney
forming complex patterns of nephropathy. Even though there are
strong genetic determinants in the risk of nephropathy, only 40–50% Hemodynamic pathways
of patients with type 1 or type 2 DM will eventually end up in neph-
ropathy.8 The strategies to determine the genetic loci for DN suscep- Glomerular hemodynamic changes occurs very early in DN, which
tibility are genomic screening and polymorphism of candidate gene include hyperfiltration and hyperperfusion injuries.14 There is a de-
crease in both afferent and efferent arteriolar resistance which is more
*Corresponding author. on the afferent side leading to increased glomerular capillary pressure
E-mail address: chinthavinod@sify.com that enhances trans-capillary hydraulic pressure gradient as well as
ISSN: 2211-9477 Copyright © 2012. Reed Elsevier India Pvt. Ltd. All rights reserved.
doi: 10.1016/S2211-9477(12)70005-5
122 P.B. Vinod / Clinical Queries: Nephrology 0102 (2012) 121–126

Table 1
Metabolic pathway Hemodynamic pathways
Renal structural abnormalities found in diabetic nephropathy.

● Mesangial expansion
Hyperglycemia High pressure ● Glomerulosclerosis (diffuse, nodular)
• AGE • RAAS ● Fibrin cap lesion
• PKC pathway • VEGF ● Capsular drop lesion
• Polyol pathway • TGF-β ● Basement membrane thickening (glomerular and tubular)
• Oxidative stress • Endothelin ● Endothelial foam cells podocyte abnormalities
● Armanni-Ebstein cells (proximal tubules stuffed with glycogen)
● Tubular atrophy
● Interstitial inflammation
Intracellular signaling molecules and ROS ● Interstitial fibrosis
● Arteriosclerosis

TGF-β, VEGF, IL-1, IL-6, IL-18, TNF-α


year. Several biochemical alterations of the GBM occur in DN.31 There
• ECM accumulation is an increase in collagen type IV deposition, whereas the expression
• GBM thickening of heparin sulfate and the extent of sulfation decreases. In contrast to
• Glomerulosclerosis the mesangial matrix in which the α1 and α2 of type IV collagen are
mainly expressed, the GBM contains α3, α4, and α5 chains.32,33 In DN,
Diabetic nephropathy there is an up-regulation of α1 (IV) and α2 (IV) chains in mesangial
cells, whereas α3 (IV) and α4 (IV) expression is increased in the GBM.33
Figure 1 Pathways involved in the development of diabetic kidney disease. AGE: Deposition of collagen type I and III in the mesangial area occurs late
advanced glycation end products, IL-1: interleukin-1, IL-6: interleukin-6, IL-18: in glomerulosclerosis and is not an early event.32
interleukin-18, PKC: protein kinase C, RAAS: renin angiotensin aldosterone system, Glomerular epithelial cells (podocytes) directly cover the GBM
ROS: reactive oxygen species, TGF-β: transforming growth factor-beta, TNF-α: tumor and there is recent evidence that alterations in structure and function
necrotic factor-alfa, VEGF: vascular endothelial growth factor.
of podocytes occur early in DN.34–36 Hyperglycemia causes changes
in integrin expression which influence the podocytes and the GBM
an increase in glomerular plasma flow.14 Hyperperfusion and hyper- interlinking process.32 Studies in human shows that there is a reduc-
filtration are also said to be due to factors such as prostanoids, nitric tion in podocyte number on follow-up, related with proteinuria.34
oxide, atrial natriuretic factor, growth hormone, glucagon, insulin and In an European collective of White type 2 diabetics, Vestra et al. found
angiotensin-II (ANG-II).14 The changes leading to glomerulosclerosis a significant reduction in the numerical density of podocytes per
are elevated intraglomerular pressure, increase in mesangial cell matrix glomerulus in patients with type 2 diabetes that were normoalbu-
production and thickening of glomerular basement membrane.15,16 minuric.37 There is a reduction in the podocyte number in diabetic
Hyperglycemia stimulate the synthesis of ANG-II, which exert hemo- patients of all ages, with reduced podocytes per glomerulus even in
dynamic, trophic, inflammatory and profibrinogenic effects on renal short duration of diabetes.38 Podocyte damage occurs before the de-
cells.15,17 The factors that mediate hyperfiltration injury include velopment of glomerulosclerosis and tubulointerstitial damage.39
vascular endothelial growth factors (VEGF) and cytokines such as
transforming growth factor-beta (TGF-β).15 The key role in diabetic
vascular derangement can be attributed to TGF-β. the mechanism Metabolic pathways
is the increase NO production by the up-regulation of endothelial
NO synthase (eNOS) mRNA expression and by enhancing arginine The glucose transport activity is an important modulator of extracel-
resynthesis.18,19 lular formation of mesangial cells. Glucose transporter-1 (GLUT-1)
Alteration of glomerular hemodynamics due to shear stress and is a key regulator of glucose entry in to kidney cells and the glucose
mechanical strain, induce the autocrine and/or paracrine release of activates various metabolic pathways leading to mesangial expansion
cytokines and growth factors.16 Hemodynamic stress causes structural and mesangial cell matrix production, mesangial cell apoptosis and
changes of DN by the local activation of cytokines and growth fac- structural changes.40 If there is overexpression of GLUT-1, similar
tors. Increase in reabsorption of sodium chloride in proximal tubules changes will be induced in a renal cell even if the glucose levels is
or loops of Henle leads to an increase in the glomerular filtration rate normal.41,45 Mesangial cells express insulin-sensitive extracellular glu-
by an intact macula-densa mechanism and hypertrophy of tubules cose transporters (GLUT-4) as well as a brain type of glucose trans-
that mediate stimulated sodium chloride reabsorption could be piv- porters (GLUT-1) through which excessive glucose could easily enter
otal in this process, linking again structural changes with hemody- the cell in an insulin-independent manner.43,44
namic adaptation in DN.20 Non-enzymatic glycosylation that produce advanced glycosylation
end products (AGE), activation of protein kinase C (PKC), and accel-
eration of the polyol pathway along with hemodynamic changes re-
Pathological changes sults in the activation of VEGF, TGF-β, interleukin-1 (IL-1), IL-6 and
IL-18 and tumor necrosis factor alpha (TNF-α). All these pathways
The hallmark of DN is nodular glomerulosclerosis and described by act in unison leading to increased albumin permeability in GBM and
Kimmelstiel and Wilson. Diabetes mellitus causes injury of all renal extracellular matrix accumulation, resulting in increasing proteinu-
compartments (Table 1) such as glomerulosclerosis, vascular diseases ria, glomerulosclerosis and finally tubulointerstitial fibrosis.
and changes of the tubulointerstitium with tubular atrophy and in-
terstitial fibrosis.21–25
Increase in extracellular matrix and mesangial cell hypertrophy Oxidative stress
causes expansion of the mesangial area which is the earliest mor-
phological change of DN.23 Mesangial cell proliferation comes to us a Increase in oxidative stress and the overproduction of reactive oxy-
standstill in the G1-phase of the cell cycle by P 27Kip1 due to the ef- gen species (ROS) in diabetes is occurring due to hyperglycemia. This
fect of hyperglycemia. The ANG-II also enhances P 27Kip 1 which is the ROS induces peroxidation of cell membrane lipids, oxidation of pro-
mediator of G1-phase arrest. Blockade of ANG-II increases glucose teins, renal vasoconstriction and deoxyribonucleic acid (DNA) dam-
mediated mesangial injury.26–31 age. Various biochemical pathways are also stimulated through the
Glomerular basement membrane (GBM) thickening starts as early increased generation of ROS mainly PKC pathways, AGE formation,
as 1 year after onset of type 1 diabetes31 which progresses over the TGF-β, and ANG-II.
P.B. Vinod / Clinical Queries: Nephrology 0102 (2012) 121–126 123

Hyperglycemia PKC. Inhibition of PKC-β, the major isoform-induced in the kidney by


hyperglycemia, ameliorates DN. Moreover, activation of PKC could,
in turn, further stimulate MAPKs. Erk 1, 2 as well as p38 MAPK have
↑ Mitochondrial ROS been implicated as signaling intermediates in DN.43 MAPKs are ad-
ditionally activated by ROS and there is likely cross-talk between the
various pathways.44 The importance of PKC in the development of
↑ Oxidative stress some changes of DN is underscored by recent studies demonstrating
that albuminuria was absent in diabetic PKC-α knockout mice.50
However, glomerular hypertrophy or the up-regulation of TGF-β was
↑ Protein kinase C not influenced by the lack of PKC-α.50

↑ Sorbitol
Advanced glycation end products
↑ Prostanoids In longstanding hyperglycemia, the excess glucose combines with
free aminoacids or tissue proteins. This glycosylation leads to the
↑ Cytokines development of DN. This process initially forms reversible early gly-
cosylation products and later irreversible AGE. The matrix proteins in
↑ AGEs the glomerular epithelial cells get accumulated along with decrease
in collagenase activity and defect in the glomerular epithelial cell
tight junction, because of the increase in AGEs.51
NF + κB

Figure 2 Reactive oxygen species as a common mediator of pathophysiological ef- Cytokines and growth factors
fects of hyperglycemia. Increased uptake of glucose into cells leads to stimulated mi-
tochondrial reactive oxygen species formation. This oxidative stress, in turn, activates
different processes involving protein kinase C, NF-κB, cytokines, formation of ad- Various growth factors, cytokines, chemokines, and vasoactive agents
vanced glycation end products, and others. AGE: advanced glycation end products, have been implicated in structural changes of DN.52–56 Insulin-like
NF: nuclear factor, ROS: reactive oxygen species. growth factors (IGFs) are among the most widely and earliest studied
growth factors in DN but the exact role remains elusive.57 An early
and temporary increase in renal IGF-I protein after the onset of dia-
betes is found in various animal models and this increase is caused
Histopathology of a diabetic kidney specimen shows accumulated by hyperglycemia.57 Interference with the IGF-I axis partly attenuates
products of glycooxidation and lipooxidation in the expanded mesang- DN in some models.52
ial matrix and nodular lesion.
Hyperglycemia results in an increase in mitochondrial ROS for-
mation.42 An increase in glucose uptake leads to overproduction of Transforming growth factor-b references changes
electron donors (NADH and FADH 2) from stimulated glycolysis and
the tricarboxylic acid cycle.42 At the mitochondrial inner membrane, Transforming growth factor-β is a profibrotic growth factor causing
where the electron transport chain is localized, the increase in elec- the expansion of mesangial matrix and renal hypertrophy in DN.58
tron donors (NADH, FADH 2) generates a high membrane potential High levels of TGF-B have been measured in the glomeruli of strpto-
by pumping protons across the inner membrane. As a consequence, zotocin diabetic rats.59 It was reported that neutralizing TGF-β anti-
electron transport is inhibited at complex III increasing the half-life body prevented diabetic renal atrophy, mesangial matrix expansion,
of free-radical intermediates of coenzyme Q, which finally reduces O2 and the development of renal failure in type 2 db/db mice.60 Connec-
to superoxide. tive tissue growth factor and heat shock proteins, which are encoded
by TGF-β, have fibrogenic effects on the kidneys of patients with dia-
betes. However, the profibrogenic actions of TGF-β1 are countered
Polyol pathways by the decreased expression of renal bone morphogenic protein 7.61
Mechanical stretch induces both gene and protein expression of TGF-
Conversion of glucose to sorbitol by aldose reductase and then to β1.62 Stretch, via the intracellular signaling molecule protein kinase C,
fructose by sorbitol dehydrogenase is occurring in the polyol pathway. causes early activation of p38 mitogen-activated protein kinase, which
An increased glucose uptake into the cell causes entry of more glucose induces TGF-β1 and fibronectin production.63 The TGF-β1 contributes
into the polyol pathway. Reduction of glucose to sorbitol requires to the cellular hypertrophy and increased synthesis of collagen,
NADPH-depleting cells of an important substrate for the regeneration which inturn leads to DN.64,65 The platelet derived growth factor-beta
of glutathione which exacerebrates intracellular oxidative stress. Three- (PDGF-β) cause histological alterations in the glomerulus. Hypergly-
deoxyglucone which is intermediate, is a precursor of AGEs.43–47 cemia up-regulates PDGF-β growth factor and its receptor in the
mesangial cells leading to enhanced TGF-β expression.66

Protein kinase C pathways


Vascular endothelial growth factor
An increased flux of glucose through the hexosamine pathway has
also been linked to mechanisms of DN, particularly an increase in Overexpression of VEGF induces diabetic renal disease by increasing
TGF-β.48 Fructose-6-phosphate from glycolysis is converted to glu- the permeability of vascular endothelium, endothelial cell prolifera-
cosamine-6-phosphate in this pathway. Glycosylation of a transcrip- tion and migration, reducing transendothelial electrical resistance, and
tion factor such as Sp1 by N-acetylglucosamine stimulates TGF-β activation of matrix-degrading protease.67,68 Synthesis of endothelial
transcription. In addition, an increase in flux through the hexosamine nitric oxide which causes vasodilatation and hyperfiltration is in-
pathway up-regulates the expression of up-stream stimulatory fac- duced by hyperglycemia, TGF-β1, ANG-II, and VEGF overexpression.
tors (USFs) which transactivate the TGF-β1 promoter.49 Intracellular The VEGF overexpression causes increased production of collagen chain
accumulation of glucose also increases de novo formation of diacylg- contributing to the increased thickening of GBM DN. Some studies
lycerol (DAG) from glycolytic intermediates such as dihydroxyace- refute causative role for high VEGF levels in DN. Instead, results imply
tone phosphate.43 An increase in DAG activates several isoforms of that low levels are harmful. Baelde et al showed that VEGF messenger
124 P.B. Vinod / Clinical Queries: Nephrology 0102 (2012) 121–126

ribonucleic acid (RNA) concentrations were decreased in the glomeruli High glucose
of patients with DN and correlated with reduction in the number of
podocytes and progression of renal disease.69
ROS
Intracellular signal pathways
AGEs ANG-II Proteinuria
Nuclear factor-kB
Nuclear factor-κB plays an important role in cell survival and its inhi-
bition leads to apoptosis. Increased monocyte NF-κB activity seen in
diabetics with nephropathy than diabetics without nephropathy.70
In vitro studies have demonstrated that high glucose, AGEs, AGN II, Cytokines (TGF-β, VEGF, MCP-1)
and stretch potently induce NF-κB activation mainly via formation of
ROS and activation of PKC71–73 providing potential cellular mecha- Inflammation
nisms of NF-κB activation in the diabetic kidney. Recent studies have
shown that NF-κB mediates both stretch and high glucose-induced
Tissue destruction fibrosis
monocyte hemoattractant protein (MCP) production in mesangial
cells73,74 playing a role in glomerular epithelial cell apoptosis75 and
Figure 3 Summary of mediators involved in the pathophysiology of diabetic nephro-
modulates the TGF-β1 intracellular signaling pathway.76 There is thus
pathy. AGE: advanced glycation end products, ANG-II: angiotensin-II, MCP: monocyte
preliminary evidence for a role of NFκB in the pathogenesis of both hemoattractant protein, ROS: reactive oxygen species, TGF-β: transforming growth
glomerular and tubular damage in diabetes. Both ACE-inhibitor and factor-beta, VEGF: vascular endothelial growth factor.
statins are potent NF-κB inhibitors, and their renoprotective action
may be, at least in part, related to the suppression of NF-κB activity.
The proinflammatory transcription factor NF-B was detected mainly
in tubular cells in biopsy specimens from 11 patients with type 2 dia-
Peroxisome proliferator-activated receptor-g betes and overt nephropathy, indicating that proteinuria may have
contributed to this activation.94 Proteases released by the mononu-
Peroxisome proliferator-activated receptor-γ (PPAR-γ) is a nuclear clear cells and cytokine such as TGF-β, leads to permanent damage to
transcription factor and the pharmacologic target for the insulin sen- nephron in DN. The ANG-II also induced the production of proinflam-
sitizers known as thiazolidinediones (TZDs). Preliminary studies sug- matory cytokines.87
gest that TZDs may ameliorate urinary albumin excretion in type 2
diabetic patients with microalbuminuria.77 Moreover, TZDs have an-
tiproteinuric effects independent of their insulin-sensitizing action Role of the renin–angiotensin–aldosterone system
in rats with streptozotocin-induced diabetes.78,79 The underlying mech-
anism of this protective action is still unclear. In diabetic glomeruli Zatz et al have shown that ACE-inhibitor prevented the development
and mesangial cells exposed to high glucose there is a strong down- of nephropathy in experimentally induced diabetes.92 Inhibition of
regulation of PPAR-γ80 and PPAR-γ ligands may counterbalance a renin–angiotensin–aldosterone system (RAAS) causes normalization
PPAR-γ relative deficit in diabetes. Furthermore, recent in vitro stud- of systemic and glomerular hypertension and also anti-fibrotic and
ies have shown that PPAR-γ ligands prevent production of both TGF- anti-inflammatory actions (Figure 3).93 The increase in local ANG-II
β1-induced collagen type 180 and stretch-induced MCP-1, providing a concentrations mediated by the hyperglycemia-induced overexpres-
potential link between PPAR-γ deficiency and glomerulosclerosis. sion of rennin and angiotensinogen in mesangial and tubular cells
which may in turn induce the production of variety of cytokines and
growth factors.93–97 Inhibition of the RAAS reduces proteinuria in DN.
Hypoxia and diabetic nephropathy Nehrin expression in podocytes suppressed by an increase in ANG-II.
The proteinuria and trasit of proteins through the ultrafiltration
In patients with diabetes nephropathy, anemia accelerates the pro- barrier and also mechanical stretch stimulates ANG-II formations in
gression of pathophysiology.81 Early treatment with erythropoietin podocytes.98,99 Chymase, an ANG-II-forming enzyme not inhibited
slows down the renal disease progression.82–83 Renal hypoxia result- by ACE inhibitors, is up-regulated in glomeruli of patients with ne-
ing from anemia aggravates interstitial fibrosis by the stimulation of phropathy owing to type 2 diabetes.100 The suppression of nephrin
factors like TGF-β and VEGF.84 Hypoxia-inducing factor-1 (HIF-1), and expression and the resulting enhanced ultrafiltration of proteins
ANG-II also plays a major role in this.85 can be attributed to the local increase in ANG-II concentration in
the podocytes.
It is seen that ANG-II increases tubular reabsorption of proteins
Inflammation and diabetic nephropathy leading to tubular inflammation and fibrosis. Aldosterone also has a
role in the development of DN. The aldosterone antagonist spironol-
Inflammatory cells like mononuclear cell infiltrate are often found actone caused increased collagen deposition in rats 3 weeks after
in the glomerular and tubular compartments on biopsy.86,87 Hyper- streptozotocin administration. The TGF-β1 expression was also sup-
glycemia increases expression of MCP-1 in mesangial cells and RANTES pressed by spiranolactone this model.93–97 These findings have been
and MCP-1 in tubule.87–91 extended to patients. In a preliminary study, Schjoedt and colleagues
High glucose leads to the generation of ROS and increases AGE observed that an increase in the plasma aldosterone level during
synthesis. Reactive oxygen species stimulate local ANG-II generation, long-term treatment with an AT1-receptor blocker (aldosterone es-
which increases proteinuria, but proteinuria also further enhances cape phenomenon) is associated with a decline in glomerular filtra-
tubular ANG-II synthesis. Angiotensin-II also increases AGE formation tion rate in patients with nephropathy owing to type 1 diabetes. These
and is pivotal in the induction of various cytokines and growth fac- data demonstrate that aldosterone contributes to the progression
tors. Some of these cytokines directly stimulate extracellular matrix of DN despite blockade of the AT1-receptor.101 On the other hand,
synthesis (e.g., TGF-β, CTGF), whereas other mediate inflammation spironolactone decreases proteinuria in patients with 2 diabetes and
(MCP-1). As a consequence of these processes, tissue destruction and early nephropathy.102 The newer antagonist of aldosterone eplerenone
fibrosis results.92,93 reduces microalbuminuria in diabetes.103 the progression to full blown
The proteinuria in combination with hyperglycemia and AGEs in- DN can be prevented by timely treatment with drugs interfering
creases the expression of chemokines in podocytes and tubular cells. with the RAAS.
P.B. Vinod / Clinical Queries: Nephrology 0102 (2012) 121–126 125

Conclusion 26. Wolf G, Ziyadeh FN. Molecular mechanisms of diabetic hypertrophy. Kidney Int
1999;56:393–405.
27. Wolf G, Schroeder R, Ziyadeh FN, Thaiss F, Zahner G, Stahl RAK. High glucose
Diabetic nephropathy develops due to the combined action of both stimulates expression of p27Kip1 in cultured mouse mesangial cells: relation-
hemodynamic and metabolic pathways. Metabolic pathways are also ship to hypertrophy. Am J Physiol 1997;273(3 Pt 2):F348–56.
activated within the diabetic kidney and result in accumulation of 28. Wolf G, Wenzel U, Ziyadeh FN, Stahl RAK. Angiotensin converting-enzyme in-
AGEs, activation of PKC, renal polyol formation and enhanced oxida- hibitor treatment reduces glomerular p16INK4 and p27Kip1 expression in dia-
betic BBdp rats. Diabetologia 1999;42:1425–32.
tive stress. These derangements activate various cytokines and growth 29. Wolf G, Schroeder R, Zahner G, Stahl RAK, Shankland SJ. High glucose-induced
factors. These mechanisms ultimately lead to renal histologic changes hypertrophy of mesangial cells requires p27Kip1, an inhibitor of cyclin-depend-
in the glomeruli in DN: mesangial expansion, GBM thickening; and ent kinases. Am J Pathol 2001;158:1091–100.
glomerular sclerosis. 30. Wolf G, Reinking R, Zahner G, Stahl RAK, Shankland SJ. Erk 1, 2 phosphorylates
p27 (Kip1): functional evidence for a role in high glucose-induced hypertrophy
The pathophysiological mechanism of the disease is needed for the of mesangial cells. Diabetologia 2003;46:1090–9.
prevention and treatment of DKD. To improve the outcome of DN a 31. Mason RM, Wahab NA. Extracellular matrix metabolism in diabetic nephropathy.
through and scientific knowledge of the complex pathophysiological J Am Soc Nephrol 2003;14:1358–73.
aspects is needed. 32. Tsilibary EC. Microvascular basement membranes in diabetes mellitus. J Pathol
2003;200:537–46.
33. Zeisberg M, Ericksen MB, Hamano Y, Neilson EG, Ziyadeh F, Kalluri R. Differential
expression of type IV collagen isoforms in rat glomerular endothelial and me-
Acknowledgment sangial cells. Biochem Biophys Res Commun 2002;295:401–7.
34. White KE, Bilous RW, Marshall SM, et al. Podocyte number in normotensive type 1
diabetic patients with albuminuria. Diabetes 2002;51:3083–9.
The author wish to thank Dr. K.P. Chintha for helping in preparing the 35. Steffes MW, Schmidt D, McCrery R, et al. Glomerular cell number in normal sub-
manuscript and arranging references. jects and in type 1 diabetic patients. Kidney Int 2001;59:2104–13.
36. Pagtalunan ME, Miller PL, Jumping-Eagle S, et al. Podocyte loss and progressive
glomerular injury in type II diabetes. J Clin Invest 1997;99:342–8.
37. Dalla Vestra M, Masiero A, Roiter AM, Saller A, Crepaldi G, Fioretto P. Is podocyte
References injury relevant in diabetic nephropathy? Studies in patients with type 2 diabetes.
Diabetes 2003;52:1031–5.
1. USRDS: The United States Renal Data System. AMJ Kidney Dis 2003;42(Suppl): 38. Gassler N, Elger M, Kränzlin B, et al. Podocyte injury underlies the progression of
1–230. focal segmental glomerulosclerosis in the fa/fa Zucker rat. Kidney Int 2001;60:
2. Remuzzi G, Ruggenenti P, Benigni A. Understanding the nature of renal disease 106–16.
progression. Kidney Int 1997;51:2–15. 39. Coimbra TM, Janssen U, Gröne HJ, et al. Early events leading to renal injury in
3. Ritz E, Rychlik I, Locatelli F, Halimi S. End-stage renal failure in type 2 diabetes: a obese Zucker (fatty) rats with type II diabetes. Kidney Int 2000;57:167–82.
medical catastrophe of worldwide dimensions. Am J Kidney Dis 1999;34:795–808. 40. Mishra R, Emancipator SN, Kern T, Simonson MS. High glucose evokes an intrin-
4. Parving HH. Diabetic nephropathy: prevention and treatment. Kidney Int 2001; sic proapoptotic signaling pathway in mesangial cells. Kidney Int 2005;67:82–93.
60:2041–55. 41. Heilig CW, Concepcion LA, Riser BL, Freytag SO, Zhu M, Cortes P. Overexpression
5. Remuzzi G, Schieppati A, Ruggenenti P. Nephropathy in patients with type 2 dia- of glucose transporters in rat mesangial cells cultured in a normal glucose milieu
betes. N Engl J Med 2002;346:1145–51. mimics the diabetic phenotype. J Clin Invest 1995;96:1802–14.
6. Harris MI, Flegal KM, Cowie CC, et al. Prevalence of diabetes, impaired fasting 42. Brownlee M. Biochemistry and molecular cell biology of diabetic complications.
glucose, and impaired glucose tolerance in US adults. Diabetes Care 1998;21: Nature 2001;414:813–20.
518–24. 43. Haneda M, Koya D, Isono M, Kikkawa R. Overview of glucose signaling in me-
7. Wolf G, Ritz E. Diabetic nephropathy in type 2 diabetes: prevention and patient sangial cells in diabetic nephropathy. J Am Soc Nephrol 2003;14:1374–82.
management. J Am Soc Nephrol 2003;14:1396–405. 44. Heilig CW, Kreisberg JI, Freytag S, et al. Antisense GLUT-1 protects mesangial
8. Berger M, Mönks D, Wanner C, Lindner TH. Diabetic nephropathy: an inherited cells from glucose induction of GLUT-1 and fibronectin expression. Am J Physiol
diseases or just a diabetic complication? Kidney Blood Press Res 2003;26:143–54. 2001;280:F657–66.
9. Rudofsky G Jr, Isermann B, Schilling T, et al. A 63 bp deletion in the promoter of 45. Heilig CW, Conception LA, Riser BL, Freytag SO, Zhu M, Cortes P. Overexpression
RAGE correlates with a decreased risk for nephropathy in patients with type 2 of glucose transporters in rat mesangial cells cultured in a normal glucose
diabetes. Exp Clin Endocrinol Diabetes 2004;112:135–41. milieu mimics the diabetic phenotype. J Clin Invest 1995;96:1802–14.
10. Hansen TK, Tarnow L, Thiel S, et al. Association between mannose-binding lectin 46. Morrison J, Knoll K, Hessner MJ, Liang M. Effect of high glucose on gene expres-
and vascular complications in type 1 diabetes. Diabetes 2004;53:1570–6. sion in mesangial cells: upregulation of the thiol pathway is an adaptational
11. Susztak K, Sharma K, Schiffer M, McCue P, Ciccone E, Böttinger EP. Genomic strat- response. Physiol Genomics 2004;17:271–82.
egies for diabetic nephropathy. J Am Soc Nephrol 2003;14:S271–8. 47. Bernobich E, Cosenzi A, Campa C, et al. Antihypertensive treatment and
12. Bowden DW, Colicigno CJ, Langefeld CD, et al. A genome scan for diabetic neph- renal damage: amlodipine exerts protective effect through the polyol pathway.
ropathy in African Americans. Kidney Int 2004;66:1517–26. J Cardiovasc Pharmacol 2004;44:401–6.
13. Imperatore G, Hanson RL, Pettitt DJ, Kobes S, Bennett PH, Knowler WC. Sib-pair 48. Kolm-Litty V, Sauer U, Nerlich A, Lehmann R, Schleicher ED. High glucose-induced
linkage analysis for susceptibility genes for microvascular complications among transforming growth factor beta1 production is mediated by the hexosamine
Pima Indians with type 2 diabetes. Diabetes 1998;47:821–30. pathway in porcine glomerular mesangial cells. J Clin Invest 1998;101:160–9.
14. Hostetter TH. Hyperfiltration and glomerulosclerosis. Semin Nephrol 2003;23: 49. Weigert C, Brodbeck K, Sawadogo M, Häring HU, Schleicher ED. Upstream stimu-
194–9. latory factor (USF) proteins induce human TGF-beta 1 gene activation via the
15. Wolf G, Butzmann U, Wenzel UO. The renin-angiotensin system and progression glucose-response element-1013/-1002 in mesangial cells: up-regulation of USF
of renal disease: from hemodynamics to cell biology. Nephron Physiol 2003;93: activity by the hexosamine biosynthetic pathway. J Biol Chem 2004;279:15908–15.
P3–13. 50. Menne J, Park JK, Boehne M, et al. Diminished loss of proteoglycans and lack of
16. Anderson S, Rennke HG, Brenner BW. Therapeutic advantage of converting en- albuminuria in protein kinase C-α-deficient diabetic mice. Diabetes 2004;53:
zyme inhibitors in arresting progressive renal disease associated with systemic 2101–9.
hypertension in the rat. J Clin Invest 1986;77:1993–2000. 51. Singh AK, Mo W, Dunea G, Arruda JA. Effect of glycated proteins on the matrix of
17. Chen S, Wolf G, Ziyadeh FN. The renin-angiotensin system in diabetic nephropathy. glomerular epithelial cells. J Am Soc Nephrol 1998;9:802–10.
Contrib Nephrol 2001;135:212–21. 52. Flyvbjerg A. Putative pathophysiological role of growth factors and cytokines in
18. Sharma K, Deelman L, Madesh M, et al. Involvement of transforming growth factor- experimental diabetic kidney disease. Diabetologia 2000;43:1205–23.
beta in regulation of calcium transients in diabetic vascular smooth muscle cells. 53. Wolf G. Growth factors and the development of diabetic nephropathy. Curr Diab
Am J Physiol Renal Physiol 2003;285:F1258–70. Rep 2003;3:485–90.
19. Tsuchida K, Cronin B, Sharma K. Novel aspects of transforming growth factor- 54. Chiarelli F, Santilli F, Mohn A. Role of growth factors in the development of dia-
beta in diabetic kidney disease. Nephron 2002;92:7–21. betic complications. Horm Res 2000;53:53–67.
20. Thomson SC, Vallon V, Blantz RC. Kidney function in early diabetes: the tubular 55. Ziyadeh FN. Mediators of diabetic renal disease: the case for TGF-beta as the
hypothesis of glomerular filtration. Am J Physiol Renal Physiol 2004;286:F8–15. major mediator. J Am Soc Nephrol 2004;15(Suppl 1):S55–7.
21. Kimmelstiel P, Wilson C. Intercapillary lesions in the glomeruli of the kidney. 56. Schrijvers BF, Flyvbjerg A, de Vriese AS. The role of vascular endothelial growth
Am J Pathol 1936;12:83–98. factor (VEGF) in renal pathophysiology. Kidney Int 2004;65:2003–17.
22. Adler S. Structure-function relationships associated with extracellular matrix al- 57. Sugimoto H, Shikata K, Makino H, Ota K, Ota Z. Increased gene expression of
terations in diabetic glomerulopathy. J Am Soc Nephrol 1994;5:1165–72. insulin-like growth factor-I receptor in experimental diabetic rat glomeruli.
23. Mauer SM, Steffes MW, Ellis EN, Sutherland DER, Brown DM, Goetz FC. Structural- Nephron 1996;72:648–53.
functional relationships in diabetic nephropathy. J Clin Invest 1984;74:1143–55. 58. Sharma K, Jin Y, Guo J, Ziyadeh FN. Neutralization of TGF-β antibody attenuates
24. DeFronzo RA. Diabetic nephropathy: etiologic and therapeutic considerations. kidney hypertrophy and the enhanced extracellular matrix gene expression in
Diab Rev 1995;3:510–64. STZ-induced diabetic mice. Diabetes 1996;45:522–30.
25. Dalla Vestra M, Saller A, Bortolosco E, Mauer M, Fioretto P. Structural involve- 59. Wang SN, LaPage J, Hirschberg R. Role of glomerular ultrafiltration of growth fac-
ment in type 1 and type 2 diabetic nephropathy. Diabetes Metab 2000;26(Suppl 4): tors in progressive interstitial fibrosis in diabetic nephropathy. Kidney Int 2000;
8–14. 57:1002–14.
126 P.B. Vinod / Clinical Queries: Nephrology 0102 (2012) 121–126

60. Ziyadeh FN, Hoffman BB, Han DC, et al. Long-term prevention of renal insuffi- 82. Gouva C, Nikolopoulos P, Ioannidis JP, Siamopoulos KC. Treating anemia early in
ciency, excess matrix gene expression, and glomerular mesangial matrix expansion renal failure patients slows the decline of renal function: a randomized control-
by treatment with monoclonal antitransforming growth factor-beta antibody in led trial. Kidney Int 2004;66:753–60.
db/db diabetic mice. Proc Natl Acad Sci USA 2000;97:8015–20. 83. Bahlmann FH, Song R, Boehm SM, et al. Low-dose therapy with the long-acting
61. Sharma K, Eltayeb BO, McGowan TA, et al. Captopril-induced reduction of serum erythropoietin analogue darbepoetin alpha persistently activates endothelial
levels of transforming growth factor-β1 correlates with long-term renoprotection Akt and attenuates progressive organ failure. Circulation 2004;110:1006–12.
in insulin-dependent diabetic patients. Am J Kidney Dis 1999;34:818–23. 84. Fine LG, Bandyopadhay D, Norman JT. Is there a common mechanism for the
62. Riser BL, Cortes P, Heilig C, et al. Cyclic stretching force selectively upregulates progression of different types of renal diseases other than proteinuria? Towards
transforming growth factor-beta isoforms in cultured rat mesangial cells. Am J the unifying theme of chronic hypoxia. Kidney Int 2000;75:S22–6.
Pathol 1996;148:1915–23. 85. Wolf G, Schroeder R, Stahl RA. Angiotensin II induces hypoxia-inducible fac-
63. Gruden G, Zonca S, Hayward A, et al. Mechanical stretch-induced fibronectin and tor-1 alpha in PC 12 cells through a posttranscriptional mechanism: role of AT2
transforming growth factor-beta 1 production in human mesangial cells is p38 receptors. Am J Nephrol 2004;24:415–21 [Epub 2004 Aug 11].
mitogen-activated protein kinase-dependent. Diabetes 2000;49:655–61. 86. Noronha IL, Fujihara CK, Zatz R. The inflammatory component in progressive
64. Janssen B, Hohenadel D, Brinkkoetter P, et al. Carnosine as a protective factor in renal disease-are interventions possible? Nephrol Dial Transplant 2002;17:
diabetic nephropathy: association with a leucine repeat of the carnosinase gene 363–8.
CNDP1. Diabetes 2005;54:2320–27. 87. Mezzano S, Droguett A, Burgos ME, et al. Renin-angiotensin system activation
65. Isaka Y, Akagi Y, Ando Y, Imai E. Application of gene therapy to diabetic neph- and interstitial inflammation in human diabetic nephropathy. Kidney Int Suppl
ropathy. Kidney Int Suppl 1997;60:S100–3. 2003;86(Suppl):S64–70.
66. Di Paolo S, Gesualdo L, Ranieri E, Grandaliano G, Schena FP. High glucose concen- 88. Lee FT, Cao Z, Long DM, et al. Interactions between angiotensin II and NF-κB-
tration induces the overexpression of transforming growth factor-beta through dependent pathways in modulating macrophage infiltration in experimental
the activation of a platelet-derived growth factor loop in human mesangial cells. diabetic nephropathy. J Am Soc Nephrol 2004;15:2139–51.
Am J Pathol 1996;149:2095–06. 89. Mezzano S, Aros C, Droguett A, et al. NF-kappaB activation and overexpression
67. Satchell SC, Anderson KL, Mathieson PW. Angiopoietin 1 and vascular endothe- of regulated genes in human diabetic nephropathy. Nephrol Dial Transplant
lial growth factor modulate human glomerular endothelial cell barrier proper- 2004;19:2505–12.
ties. J Am Soc Nephrol 2004;15:566–74. 90. Kikuchi Y, Ikee R, Hemmi N, et al. Fractalkine and its receptor, CX3CR1, upregu-
68. Kanesaki Y, Suzuki D, Uehara G, et al. Vascular endothelial growth factor gene lation in streptozotocin-induced diabetic kidneys. Nephron Exp Nephrol 2004;97:
expression is correlated with glomerular neovascularization in human diabetic e17–25.
nephropathy. Am J Kidney Dis 2005;45:288–94. 91. Utimura R, Fujihara CK, Mattar AL, Malheiros DM, Noronha IL, Zatz R. Mycophe-
69. Baelde HJ, Eikmans M, Lappin DW, et al. Reduction of VEGF-A and CTGF expres- nolate mofetil prevents the development of glomerular injury in experimental
sion in diabetic nephropathy is associated with podocyte loss. Kidney Int 2007; diabetes. Kidney Int 2003;63:209–16.
71:637–45. 92. Zatz R, Dunn BR, Meyer TW, Anderson S, Rennke HG, Brenner BM. Prevention of
70. Hofmann MA, Schiekofer S, Isermann B, et al. Peripheral blood mononuclear cells diabetic glomerulopathy by pharmacological amelioration of glomerular capil-
isolated from patients with diabetic nephropathy show increased activation of the lary hypertension. J Clin Invest 1986;77:1925–30.
oxidative-stress sensitive transcription factor NF-κB. Diabetologia 1999;42:222–32. 93. Wolf G, Ziyadeh FN. The role of angiotensin II in diabetic nephropathy: empha-
71. Pieper GM, Riaz-ul-Haq. Activation of NFκB in cultured endothelial cells by in- sis on nonhemodynamic mechanisms. Am J Kidney Dis 1997;29:153–63.
creased glucose concentration: prevention by calphostin C. J Cardiovasc Pharmacol 94. Vidotti DB, Casarini DE, Cristovam PC, Leite CA, Schor N, Boim MA. High glucose
1997;30:528–32. concentration stimulates intracellular renin activity and angiotensin II genera-
72. Lal MA, Brismar H, Eklöf AC, Aperia A. Role of oxidative stress in advanced glyca- tion in rat mesangial cells. Am J Physiol Renal Physiol 2004;286:F1039–45.
tion end product-induced mesangial cell activation. Kidney Int 2002;61:2006–14. 95. Singh R, Sing AK, Alavi N, Leehey DJ. Mechanism of increased angiotensin II lev-
73. Ha H, Yu MR, Choi YJ, Kitamura M, Lee HB. Role of high glucose-induced nuclear els in glomerular mesangial cells cultured in high glucose. J Am Soc Nephrol
factor-kappaB activation in monocyte chemoattractant protein-1 expression by 2003;14:873–80.
mesangial cells. J Am Soc Nephrol 2002;13:894–902. 96. Hsieh TJ, Zhang SL, Filep JG, Tang SS, Ingelfinger JR, Chan JS. High glucose stimu-
74. Setti G, Gruden G, Hayward A, et al. Rosiglitazone prevents stretch-induced lates angiotensinogen gene expression via reactive oxygen species generation
monocyte recruitment by inhibiting NFKB-MCP-1 pathway in human mesangial in rat kidney proximal tubular cells. Endocrinology 2002;143:2975–86.
cells. Diabetic Med 2003;20(Suppl 2):A7. 97. Zhang SL, To C, Chen X, et al. Essential roles (s) of the intrarenal renin-angio-
75. Okado T, Terada Y, Tanaka H, Inoshita S, Nakao A, Sasaki S. Smad7 mediates trans- tensin system in transforming growth factor-β1 gene expression and induction
forming growth factor-beta-induced apoptosis in mesangial cells. Kidney Int of hypertrophy of rat kidney proximal tubular cells in high glucose. J Am Soc
2002;62:1178–86. Nephrol 2002;13:302–12.
76. Nagarajan RP, Chen F, Li W, et al. Repression of TGF-β1-mediated transcription by 98. Abbate M, Zoja C, Morigi M, et al. Transforming growth factor-beta1 is upregu-
nuclear factor κB. Biochem J 2000;348:591–6. lated by podocytes in response to excess intraglomerular passage of proteins:
77. Nakamura T, Ushiyama C, Suzuki S, et al. Effect of troglitazone on urinary albu- a central pathway in progressive glomerulosclerosis. Am J Pathol 2002;161:
min excretion and serum type IV collagen concentrations in type 2 diabetic pa- 2179–93.
tients with microalbuminuria or macroalbuminuria. Diabet Med 2001;18:308–13. 99. Durvasula RV, Petermann AT, Hiromura K, et al. Activation of a local tissue angio-
78. Yamashita H, Nagai Y, Takamura T, Nohara E, Kobayashi K. Thiazolidinedione de- tensin system in podocytes by mechanical strain. Kidney Int 2004;65:30–9.
rivatives ameliorate albuminuria in streptozotocin-induced diabetic spontane- 100. Huang XR, Chen WY, Truong LD, Lan HY. Chymase is upregulated in diabetic
ous hypertensive rat. Metabolism 2002;51:403–8. nephropathy: implications for an alternative pathway of angiotensin II-mediated
79. Isshiki K, Haneda M, Koya D, Maeda S, Sugimoto T, Kikkawa R. Thiazolidinedione diabetic renal and vascular disease. J Am Soc Nephrol 2003;14:1738–47.
compounds ameliorate glomerular dysfunction independent of their insulin- 101. Schjoedt KJ, Andersen S, Rossing P, Tranow L, Parving HH. Aldosterone escape
sensitizing action in diabetic rats. Diabetes 2000;49:1022–32. during angiotensin II receptor blockade in diabetic nephropathy is associated
80. Zheng F, Fornoni A, Elliot SJ, et al. Upregulation of type I collagen by TGF-β in with enhanced decline in GFR (Abstract). J Am Soc Nephrol 2003;14:7A.
mesangial cells is blocked by PPARγ activation. Am J Physiol 2002;282:F639–48. 102. Sato A, Hayashi K, Naruse M, Saruta T. Effectiveness of aldosterone blockade in
81. Mohanram A, Zhang Z, Shahinfar S, Keane WF, Brenner BM, Toto RD. Anemia and patients with diabetic nephropathy. Hypertension 2003;41:64–8.
end-stage renal disease in patients with type 2 diabetes and nephropathy. Kidney 103. Hollenberg NK. Aldosterone in the development and progression of renal
Int 2004;66:1131–8. injury. Kidney Int 2004;66:1–9.

You might also like