Вы находитесь на странице: 1из 40

0163-769X/04/$20.00/0 Printed in U.S.A.

Endocrine Reviews 25(6):9711010 Copyright 2004 by The Endocrine Society doi: 10.1210/er.2003-0018

From Hyperglycemia to Diabetic Kidney Disease: The Role of Metabolic, Hemodynamic, Intracellular Factors and Growth Factors/Cytokines
BIEKE F. SCHRIJVERS, AN S. DE VRIESE,
AND

ALLAN FLYVBJERG

Medical Department M (B.F.S., A.F.), Medical Research Laboratories, Institute of Experimental Clinical Research, Aarhus University Hospital, DK-8000 Aarhus, Denmark; and Renal Unit (B.F.S., A.S.D.V.), Department of Internal Medicine, Gent University Hospital, B-9000 Gent, Belgium
At present, diabetic kidney disease affects about 1525% of type 1 and 30 40% of type 2 diabetic patients. Several decades of extensive research has elucidated various pathways to be implicated in the development of diabetic kidney disease. This review focuses on the metabolic factors beyond blood glucose that are involved in the pathogenesis of diabetic kidney disease, i.e., advanced glycation end-products and the aldose reductase system. Furthermore, the contribution of hemodynamic factors, the renin-angiotensin system, the endothelin system, and the nitric oxide system, as well as the prominent role of the intracellular signaling molecule protein kinase C are discussed. Finally, the respective roles of TGF-, GH and IGFs, vascular endothelial growth factor, and plateletderived growth factor are covered. The complex interplay between these different pathways will be highlighted. A brief introduction to each system and description of its expression in the normal kidney is followed by in vitro, experimental, and clinical evidence addressing the role of the system in diabetic kidney disease. Finally, well-known and potential therapeutic strategies targeting each system are discussed, ending with an overall conclusion. (Endocrine Reviews 25: 9711010, 2004)

I. Introduction II. Metabolic Factors A. Advanced glycation endproducts (AGEs) B. Aldose reductase (AR) /polyol pathway III. Hemodynamic Factors A. Angiotensin II (Ang II)/renin-angiotensin system (RAS) B. Endothelin (ET) C. Nitric oxide (NO) IV. Intracellular Factors A. Diacylglycerol (DAG)-protein kinase C (PKC) pathway V. Growth Factors and Cytokines A. Transforming growth factor (TGF-) B. Growth hormone (GH) and insulin-like growth factors (IGFs) C. Vascular endothelial growth factor (VEGF) D. Platelet-derived growth factor (PDGF) VI. Outlook and Future Perspectives
Abbreviations: ACE, Angiotensin-converting enzyme; ACEi, ACE inhibitor(s); ADMA, asymmetric dimethylarginine; AGE, advanced glycation endproduct; AGE-R, AGE receptor; AGE-R3, AGE-R type 3, galectin-3; AKR, aldo-keto reductase(s); Ang, angiotensin; AR, aldose reductase; ARB, Ang receptor blocker; ARI, AR inhibitor; AS-ODN, antisense oligodeoxynucleotide; AT1, Ang II type 1 receptor; BB, BioBreeding; bFGF, basic fibroblast growth factor; BMT, basement membrane thickness; cGMP, cyclic GMP; CML, carboxymethyl lysine; CrCl, creatinine clearance; CTGF, connective tissue growth factor; DAG, diacylglycerol; DDAH, dimethylarginine-dimethylaminohydrolase; ECE, ET-converting enzyme; ECM, extracellular matrix; EM, electron microscopic; ET, endothelin; ETA, ET receptor A; ETB, ET receptor B; GFR, glomerular filtration rate; GHBP, GH binding protein; GHR, GH receptor; I/D polymorphism, insertion/deletion polymorphism; IGFBP, IGF binding protein; IGFBP-rP, IGFBP-related proteins; IGF-II/man-6-PR,

I. Introduction

IABETIC NEPHROPATHY IS one of the most serious complications of diabetes and the most common cause of end-stage renal failure in the Western world. At present, diabetic kidney disease affects about 15 to 25% of type 1 diabetic patients (1) and 30 to 40% of patients with type 2 diabetes (2, 3). Diabetic nephropathy is characterized by specific renal morphological and functional alterations. Features of early diabetic renal changes are glomerular hyperfiltration, glomerular and renal hypertrophy, increased urinary albumin excretion (UAE), increased basement membrane thickness (BMT), and mesangial expansion with the accumulation of extracellular matrix (ECM) proteins such as collagen, fibronectin, and laminin. Advanced diabetic ne-

IGF-II/mannose-6-phosphate receptor; IGF-IR, IGF-I receptor; l-NAME, Ng-nitro-l-arginine methyl ester; l-NMMA, NG-monomethyl-l-arginine; L-NNA, N--nitro-l-arginine; MMP, matrix metalloproteinase; NADPH, nicotinamide adenine dinucleotide phosphate, reduced form; NF-B, nuclear factor-B; NO, nitric oxide; NOD, nonobese diabetic; NOS, NO synthase; NOS1/nNOS, neuronal NO synthase; NOS2/iNOS, inducible NO synthase; NOS3/eNOS, endothelial NOS; NOx, NO2 and NO3; OLETF, Otsuka-Long-Evans-Tokushima-fatty; PDGF, platelet-derived growth factor; PDGFR, PDGF receptor; PKC, protein kinase C; PM, pyridoxamine dihydrochloride; PTB, N-phenacetylthiazolium bromide; RAGE, receptor for AGEs; RAS, renin-angiotensin system; RPF, renal plasma flow; ScR-II, macrophage scavenger receptor type II; SDH, sorbitol dehydrogenase; SNP, single nucleotide polymorphism; SOD, superoxide dismutase; SPARC, secreted protein acidic and rich in cysteine; STZ, streptozotocin; TGF- RI, TGF- receptor type I; TKi, tyrosine kinase inhibitor(s); TZD, thiazolidinedione; UAE, urinary albumin excretion; VEGF, vascular endothelial growth factor; VSMC, vascular smooth muscle cell; ZDF, Zucker diabetic fatty. Endocrine Reviews is published bimonthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.

971
Downloaded from edrv.endojournals.org on April 19, 2006

972

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

phropathy is characterized by proteinuria, a decline in renal function, decreasing creatinine clearance (CrCl), glomerulosclerosis, and interstitial fibrosis. As will appear from this review, a vast majority of the studies referenced have been performed in diabetic animals. This raises the question whether diabetic rodents are good models of human disease. The most frequently used animal strain in experimental diabetes is the rat. In spontaneous and chemically [e.g., streptozotocin (STZ)] induced rat models of type 1 diabetes, as well as rat models of type 2 diabetes, early renal changes are seen: renal enlargement, glomerular hypertrophy, hyperfiltration, and an increasing UAE (4, 5). Furthermore, diabetic rats with a diabetes duration of more than 6 months develop electron microscopic (EM) changes, i.e., increased BMT (6 8). Over the past years, diabetic mice models have been used more frequently in the study of diabetic renal changes. As is the case for rats, diabetic mice present with early renal changes comparable with the changes seen in diabetic rats, i.e., renal enlargement, glomerular hypertrophy, hyperfiltration, and a progressive increase in UAE (9). In contrast to diabetic rats, diabetic mice models develop pronounced structural glomerular EM changes after approximately 2 months duration of diabetes with an increase in BMT and mesangial expansion (9, 10). In conclusion, various diabetic rodent models display early renal changes that have similarities to the early changes seen in human diabetes, whereas only diabetic mice present with increased BMT and mesangial expansion at an EM-level within a diabetes duration of a few months. Over the past few decades, extensive research has elucidated several pathways that play a role in the development and/or progression of diabetic kidney disease. Beyond the role of high blood glucose, the pathophysiological role of different metabolic pathways in the development and progression of diabetic nephropathy has been studied extensively. Advanced glycation endproduct (AGE) formation as a consequence of sustained or transient hyperglycemia has been implicated. Furthermore, the potential role of the aldose reductase/polyol pathway, although controversial, will be addressed. In addition, various vasoactive factors contribute to the development and progression of diabetic microvascular complications. These vasoactive factors include vasoconstrictors such as angiotensin (Ang) II and endothelin (ET), as well as vasodilators such as nitric oxide (NO). Besides the well-known systemic and locally hemodynamic effects, the renin-angiotensin system (RAS) and the ET system can exert nonhemodynamic effects via an autocrine or paracrine mode of action. They stimulate the proliferation of kidney cells and the expression of growth factors or cytokines, which may directly or indirectly contribute to the renal changes seen in diabetes. The controversial role of the NO system in renal hemodynamic changes in diabetes will also be addressed. Activation of the diacylglycerol (DAG)-protein kinase C (PKC) pathway is also a well-known feature of diabetes. PKC is an important intracellular pathway that can be activated by many of the metabolic and hemodynamic factors involved in the pathogenesis of diabetic nephropathy. PKC can then be a stimulus for the initiation of several growth factors and cytokines.

Different growth factors and cytokines directly influence kidney function or exert their effects in a more indirect way by stimulating other factors. One of the first endocrine factors to be implicated in the pathogenesis of diabetes was GH (11). The attention was drawn to the possible role of GH in the development of diabetic microangiopathy (12). At about the same time, IGFs were identified in diabetic serum (13). Since then, new information has appeared with increasing pace on growth factors implicated in the pathogenesis of the renal functional and structural alterations seen in diabetes. Ample experimental and clinical studies illustrate the prominent role of TGF-, and during the last several years an increasing interest has developed in vascular endothelial growth factor (VEGF). Although platelet-derived growth factor (PDGF) has been studied less intensively, evidence supporting its pathophysiological role in diabetic kidney disease has been collected.

II. Metabolic Factors A. Advanced glycation endproducts (AGEs)

1. The AGE system. When glucose and other reactive carbonyl compounds react nonenzymatically with proteins, lipids, or nucleic acids, Schiff bases and Amadori products are formed. Additional rearrangement and modification leads to the generation of diverse AGEs, such as carboxymethyl lysine (CML), pentosidine, imidazolone, and pyrraline (14, 15). AGEs interact with specific receptors (14 16). p60 (OST-48, AGE-R1), p90 (80K-H, AGE-R2), galectin-3 (AGE-R3), the macrophage scavenger receptor type II (ScR-II), and CD36 regulate the uptake and clearance of AGEs (14, 16). The best-characterized receptor is receptor for AGEs (RAGE) (14). Another molecule with AGE-binding and anti-AGE properties is the antimicrobial protein lysozyme (16). AGEs can also act in a receptor-independent way by cross-linking proteins (14, 15). AGEs alter the structure and function of intra- and extracellular molecules, increase oxidative stress, and modulate cell activation, signal transduction, and the expression of cytokines and growth factors through receptor-dependent and receptor-independent pathways (14 16). 2. Expression of AGEs in the normal kidney. In human kidney, immunostaining for pentosidine, pyrraline, CML, and imidazolone was negative in glomeruli (1719), but in renal tubuli, immunostaining was negative for CML and pentosidine in one study (19) but positive for pentosidine, CML, and imidazolone in others (17, 18). AGE staining was present in rodents, predominantly in endothelial cells within glomeruli and tubulointerstitium (20, 21). AGE and RAGE colocalize in the rat glomerulus (21). In rat kidney, RAGE is expressed in cortex and medulla (22) and localized primarily to glomeruli, distal tubules, and collecting ducts (21). In human kidney, a low level of RAGE is constitutively expressed in glomerular podocytes, but not in the mesangium or glomerular endothelium (19). In the mouse kidney, mRNA and protein for AGE-R1, AGE-R2, and AGE-R3 were detected as well as mRNA for RAGE and the ScR-II (23). More particularly, a distinct glomerular AGE-R1 staining was found, and AGE-R1 to -3 were also detected in cultured

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 973

mouse mesangial cells (23). RAGE was detected in cultured mouse podocytes (10). Human-, rat-, and mouse-cultured mesangial cells contain binding sites for AGE-modified proteins (24, 25). AGE binding sites were also identified in proximal tubules of rat kidney, but it is unclear whether they represent one of the known AGE receptors (26). 3. In vitro evidence for AGEs effects on renal cells. In cultured glomerular endothelial and mesangial cells, glycated albumin and AGE-rich proteins increased PKC activity (2729), PKC (29), TGF-1 levels (27, 30), and ECM expression (24, 25, 27, 29, 30). Hyperglycemia and glycated albumin appeared to exert an additive effect in glomerular endothelial cells (27). In cultured kidney epithelial cells, CML reduced proteoglycan expression (31). In a proximal tubular epithelial cell line, AGEs induced tubular-epithelial myofibroblast transdifferentiation through RAGE signaling (32). 4. Experimental evidence for a role of AGEs in diabetic kidney disease. In vivo evidence for a role of AGEs in diabetic kidney disease comes mainly from studies in STZ-diabetic rats. BSAAGE gold conjugate binding to mesangial matrix, BMT, and glomerular cell nuclei was more intense in renal tissue from diabetic than from control rats (33). Whether this was associated with an up-regulation of RAGE was not investigated. In STZ-diabetic rats, AGE staining was increased in the glomeruli, predominantly in glomerular endothelial cells, and tubulointerstitium (21). Renal AGE levels were increased after 3 (26), 12 (34), and 32 wk of diabetes (20, 35). Furthermore, exogenously administered AGEs induced ECM genes (36), mesangial expansion (37), and up-regulation of TGF-1 in nondiabetic mice and rats (36, 37). Recently, it was demonstrated in nonobese diabetic (NOD) and db/db mice, models of type 1 and type 2 diabetes, respectively, that the intake of food-derived AGEs contributes to diabetic nephropathy and that low AGE diet provides renoprotection (9). An increased number of proximal tubular AGE binding sites was found in type 1 diabetic rats together with an increase in renal AGE levels (26). Recently, in STZ-diabetic rats, RAGE was up-regulated after 4 wk of diabetes up to 12 wk, whereas AGE-R2 and AGE-R3 remained unchanged (22, 34). In NOD mice, early after the onset of diabetes, AGE-R1 expression was reduced, AGE-R2 expression was unchanged, and AGER3, RAGE, and ScR-II expression was increased compared with nondiabetic control mice (23). In db/db mice, glomerular RAGE staining was enhanced, especially in podocytes, compared with nondiabetic control mice (10). Diabetic transgenic mice overexpressing human RAGE developed renal and glomerular hypertrophy, increased albuminuria, mesangial expansion, advanced glomerulosclerosis, and increased serum creatinine compared with diabetic littermates lacking the RAGE transgene (38). 5. Clinical evidence for a role of AGEs in diabetic kidney disease. In type 1 diabetic patients, increased circulating AGEs preceded the development of microvascular complications (39) and predicted the progression of the early morphological renal changes in BMT and matrix/glomerular volume fraction (40). In monocytes of type 1 diabetic patients, reduced expression of AGE-R1 was linked to elevated serum AGE levels and the presence of severe diabetic complications (41).

In type 2 diabetic patients, increased serum levels of crossline, one of the structurally defined adducts of AGEs, were associated with the presence of nephropathy (42). In addition, serum concentrations of AGEs were associated with the development of diabetic microangiopathy in patients with type 2 diabetes (43). Amadori products, pentosidine, CML, pyrraline, and imidazolone were demonstrated in renal tissue of diabetic patients (17, 18). CML accumulated predominantly in the basement membrane and pentosidine primarily in the interstitium in patients with diabetic kidney disease (17, 19). The extent of CML and pentosidine immunostaining in the glomerular and tubulointerstitial compartments correlated with the severity of diabetic nephropathy (19). The distinctive pattern of CML accumulation in diabetic nephropathy differed from the more nonspecific AGE accumulation observed in nondiabetic sclerosing renal diseases. Glomeruli of patients with diabetic nephropathy demonstrated diffuse up-regulation of RAGE expression in podocytes (19). 6. Agents with effects on AGEs in diabetic kidney disease. Various classes of drugs are able to interfere with the formation of AGEs or the cross-linking of proteins by AGEs. Aminoguanidine, pyridoxamine, 2,3-diamino-phenazine, OPB-9195, and tenilsetam inhibit AGE formation by scavenging reactive carbonyl intermediates, N-phenacetylthiazolium bromide (PTB) and ALT-711 are AGE-cross-link breakers. Furthermore, signal transduction through RAGE can be inhibited by antisense oligodeoxynucleotides (AS-ODNs), RAGE antibodies, or soluble RAGE. Drugs targeting other systems have also shown some effects on AGE-related pathways. a. Glycated albumin antagonists. Treatment of db/db mice with monoclonal antibodies against Amadori-modified glycated albumin (A717) attenuated the cortical overexpression of 1 (IV) collagen and fibronectin mRNAs, ameliorated mesangial matrix expansion, reduced albuminuria, and prevented the decline in renal function that developed in untreated db/db mice (44, 45). Oral administration of EXO-226 or 23CPPA, compounds that reduce circulating glycated albumin, had similar renoprotective effects in db/db mice (46, 47). The effects of 23CPPA were mediated by a reduction in glomerular TGF-1 (47). b. Inhibitors of AGE formation. Aminoguanidine retarded the development of diabetic nephropathy in long-term experimental diabetes in rats (6, 20, 35, 48). It appears that the renoprotective effects of aminoguanidine in diabetes are related to the duration and not the timing of treatment (20, 49) and are mediated by a decrease of AGE formation (21, 26, 50). In addition, aminoguanidine restored the diabetes-induced changes in lysosomal processing (51) and glomerular PKC activity (49, 52) in STZ-diabetic rats. Recently, in STZ-diabetic transgenic (mREN-2)27 rats, a model of diabetic kidney disease with hypertension and an overactive RAS, aminoguanidine ameliorated glomerulosclerosis and medullary pathology and decreased glomerular AGE immunolabeling, but had no effect on kidney weight, glomerular filtration rate (GFR), or UAE (53). Aminoguanidine had no effect on the expression of RAGE, AGE-R2, and AGE-R3 in STZ-diabetic rats. Compared with placebo, aminoguanidine modestly re-

Downloaded from edrv.endojournals.org on April 19, 2006

974

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

duced proteinuria and marginally slowed progression of overt diabetic nephropathy in optimally treated type 1 diabetic patients (54). However, two double-blinded, placebocontrolled randomized clinical trials with aminoguanidine were performed in type 1 (I) and type 2 (II) diabetic patients with overt nephropathy. The primary endpoint, i.e., reducing the risk of doubling serum creatinine, was not achieved in ACTION I, but pimagedine reduced urinary protein excretion, serum triglycerides, and low-density lipoprotein levels. ACTION II was terminated early because of side effects and apparent lack of efficacy (55). Pyridorin (pyridoxamine dihydrochloride) (PM) inhibits the conversion of Amadori intermediates to AGEs in vitro (56). PM inhibited the increase of albuminuria and serum creatinine in STZ-diabetic rats (57). The therapeutic potential of PM is currently being investigated in humans with diabetic nephropathy (15). 2,3Diamino-phenazine inhibited AGE accumulation in vitro (50). In STZ-diabetic rats, 2,3-diamino-phenazine normalized the renal AGE staining (50) and ameliorated collagen solubility, but had no effect on increased UAE (58). OPB-9195 effectively inhibited AGE formation and AGE-derived crosslinking in vitro. In Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a model of type 2 diabetes, the administration of OPB-9195 lowered elevated serum AGE levels and UAE, attenuated glomerular AGE deposition, and prevented the progression of glomerulosclerosis (8). Furthermore, administering OPB-9195 in a RAGE transgenic mouse model prevented the features of advanced diabetic nephropathy (38). In vitro, tenilsetam restored the reduced digestibility of collagen, and in vivo, administration of tenilsetam for 16 wk suppressed the elevated levels of AGE-derived fluorescence and pyrraline in renal cortex of STZ-induced diabetic rats (59). c. AGE-receptor blockers. In cultured mesangial cells, antip60 antibodies prevented the AGE-induced increases in IGF-I, TGF-1, and ECM expression or production (60), and anti-p60 and anti-p90 antibodies prevented the AGEinduced increase in type IV collagen (24). In db/db mice treated with a neutralizing murine RAGE antibody for 2 months, the increases in kidney weight, glomerular volume, mesangial volume, and UAE were reduced, and the increases in CrCl and BMT were normalized (Fig. 1) (61). In db/db mice, treatment with soluble RAGE prevented the increases in VEGF and TGF- expression, mesangial expansion, BMT, and UAE and preserved renal function. Similarly, these di-

abetes-induced changes were not seen in diabetic RAGE null mice (10). d. AGE-cross-link breakers. PTB cleaves covalent, glucosederived protein cross-links in vitro and in vivo (62). However, PTB treatment did not affect the elevated UAE or the decreased collagen pepsin solubility in STZ-diabetic rats (58). The lack of effect could be due to the unstable nature of PTB. The stable PTB derivative ALT-711 reduced renal AGE accumulation, TGF- immunostaining, and tubular-epithelial myofibroblast transdifferentiation in STZ-diabetic rats (32). ALT-946 is a potent inhibitor of AGE accumulation and AGEprotein cross-linking (63). Treatment with ALT-946 for 16 to 32 wk attenuated the increases in GFR, albuminuria, and glomerular AGE staining observed in STZ-diabetic rats (63). Recently, in STZ-diabetic (mREN-2)27 rats, treatment with ALT-946 for 12 wk ameliorated glomerulosclerosis and medullary pathology, reduced cortical tubular degeneration, UAE, and glomerular AGE staining, but had no effect on kidney weight or GFR (53). The renoprotective effect of AGEcross-link breakers has not been tested in clinical trials. e. Aldose reductase inhibitors (ARIs). Epalrestat decreased the elevated levels of fructose 3-phosphate and AGEs in erythrocytes after 2 months of treatment (64), supporting the hypothesis that the polyol pathway plays a substantial role in the nonenzymatic glycation of proteins (64, 65). f. Others. In cultured mesangial cells, lysozyme suppressed the AGE-enhanced expression of several modulators of kidney structure and function, i.e., PDGF-B, type 1 (IV) collagen and tenascin, and normalized the AGE-suppressed expression and activity of matrix metalloproteinase-9 (MMP). In vivo, lysozyme administration to NOD and db/db mice reduced the elevated serum AGE levels, enhanced urinary AGE excretion, and decreased UAE (66). PDGF-antibody abrogated the AGE-induced increase in type IV collagen in cultured mesangial cells, indicating that this response was not mediated directly by AGEs and RAGE, but indirectly through an intermediate factor, i.e., PDGF (24). Angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme (ACE) inhibitors (ACEi) lower the formation of AGEs in vitro by lowering the production of reactive carbonyl precursors (67). Ramipril attenuated the renal AGE accumulation found in STZ-diabetic rats, identifying a relationship between the RAS and the accumulation of AGEs in experimental diabetic nephropathy (34).

FIG. 1. BMT and total mesangial volume on d 60 in placebo-treated db/ mice (open bars), RAGE antibody-treated db/db mice (light gray bars), placebo-treated db/db mice (black bars), and RAGE antibody-treated db/db mice (dark gray bars). Data are means SEM, n 6 10 in each group. *, P 0.01 vs. db/ mice and RAGE antibody-treated db/db mice. , P 0.05 vs. db/ mice and placebo-treated db/db mice. [Reproduced with permission from A. Flyvbjerg et al.: Diabetes 53:166 172, 2004 (61). American Diabetes Association.]

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 975

7. Conclusion. In vitro and experimental data suggest that the up-regulation of AGEs and RAGE, and AGE-RAGE interaction contribute to diabetic renal changes, i.e., TGF-1 upregulation, PKC activation, renal and glomerular hypertrophy, albuminuria, ECM production, mesangial expansion, and glomerulosclerosis. Reduced AGE-R1-dependent clearance may be responsible for increased circulating AGEs that are associated with the development of microvascular complications. AGEs accumulate in the kidney, especially in diabetic lesions. Inhibitors of AGE formation are promising agents because they improve functional and/or structural changes associated with diabetic kidney disease in experimental models. Despite promising preliminary effects in type 1 diabetic patients, aminoguanidine still needs to prove its effect in randomized clinical trials. Although the renoprotective effects of AGE formation inhibitors are mainly mediated through decreased AGE formation, multiple pathways are affected, i.e., cytokines such as TGF-1, IGF-I, PDGF-B, VEGF, PKC activity, and NO synthase (NOS). The renal effects of AGE-receptor blockers, RAGE antibodies, and AGE-cross-link breakers implicate AGE signaling pathways in the functional and structural changes of diabetic kidney disease.
B. Aldose reductase (AR)/polyol pathway

the papilla, in endothelial, epithelial, interstitial and collecting duct cells. In the outer medulla, it was low in proximal tubules and thick limb cells and strong in endothelial, distal tubular, and collecting duct cells (76, 77). Vascular smooth muscle cells (VSMCs) were positive for SDH, but glomeruli were all negative in control patients (77). In cultured rat mesangial cells, SDH activity was clearly detected (79). Na/ Myo-inositol cotransporter mRNA was detected predominantly in the papilla and inner medulla endothelial, epithelial, interstitial, and collecting duct cells; in the outer medulla collecting duct, endothelial, and thin limb cells; and in the cortex, Bowmans capsule, glomerular tuft, collecting duct, and distal convoluted tubule cells (76). 3. In vitro evidence for AR effects on renal cells. During hyperglycemia, glucose levels rapidly increase in tissues, such as the kidney, that are insulin-independent for glucose uptake. Excess glucose enters the polyol pathway and activates AR, but because SDH activity does not increase similarly, sorbitol accumulates (69). To explain the role of the polyol pathway in the onset of diabetic complications, different mechanisms have been proposed: accumulation of sorbitol or fructose (68, 69), myo-inositol depletion (70), or alterations in the NADPH/NADP and NADH/NAD ratios (80). In cultured rat mesangial cells, enhanced expression of the facilitative glucose transporter 1 increased AR expression and activity, polyol accumulation, and PKC levels, which may induce stimulation of matrix protein synthesis (81). In addition, in cultured mesangial cells, high glucose-induced DAG accumulation, PKC activation, and TGF- overproduction were mediated through polyol pathway activation (82, 83). In cultured proximal tubular cells, PKC and polyol pathway activation mediated the high glucose-induced collagen and fibronectin accumulation by decreasing their degradation (84 86). In proximal tubular cells, glucose increased sorbitol, fructose, DAG levels, PKC activity, and the expression of angiotensinogen (87), but not AR mRNA, immunoreactivity, and activity, which may be explained by a negative feedback of intracellular sorbitol accumulation on AR gene transcription (84, 88). 4. Experimental evidence for a role of AR in diabetic kidney disease. Both short- and long-term experimental type 1 diabetes are associated with increased renal sorbitol and fructose levels (65, 89 93). Renal myo-inositol levels were found to be increased (92, 94) or decreased in diabetic animals (95). More specifically, renal polyol accumulation was associated with a decrease in myo-inositol levels in the kidney cortex while being unchanged in the kidney medulla (93). Renal AR level and activity were elevated in diabetic rats, but renal SDH level or activity did not change (92, 96). However, one study found a decrease in glomerular AR expression in type 1 diabetic rats after 2 wk of diabetes, whereas the expression of AR did not change in glomeruli and renal arterioles of type 2 diabetic rats (97). NaK-ATPase activity was reduced in glomeruli of diabetic animal models (95). In contrast, renal NaK-ATPase activity was unaffected in another study (93). Despite the discrepancy of these results, which may be due to variations in quantification methods, duration of diabetes, degree of hyperglycemia or animal strain, the ma-

1. The AR system. AR is a member of the aldo-keto reductases (AKR), a superfamily of nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reductases with potential roles in the detoxification of aldehydes and ketones (68). The family of AR consists of 11 members, named AKR1B1 to AKR1B11 (AKR homepage, http://www.med.upenn.edu/ akr). AR catalyzes the reduction of glucose to sorbitol, the first and the rate-limiting step in the polyol pathway (68). Sorbitol metabolism is also governed by sorbitol dehydrogenase (SDH), the enzyme responsible for the degradation of sorbitol to fructose using NAD (69). Sorbitol may interfere with the uptake and metabolism of myo-inositol (70). The physiological role of the AR pathway remains largely unknown. However, AR, sorbitol and myo-inositol are thought to play a role in the osmoregulation of the kidney (71). In most mammalian cells, the intracellular concentration of myo-inositol is many times higher than in the circulation by virtue of a Na/myo-inositol cotransporter (72). Species- and organ-specific ARs have been implicated in hormone-regulated apoptosis (73) and hormone production (74). 2. Expression of AR in the normal kidney. A single form of AR is expressed in human kidney (75). AR mRNA, protein, and enzyme activity were mainly found in the inner medulla (76). AR mRNA expression was pronounced in the papilla, especially in collecting duct cells, epithelial, endothelial, interstitial, and thin limb cells, and weak in the outer medulla and cortex, particularly in cells of the collecting duct, Bowmans capsule, and the glomerular tuft (76, 77). AR protein was localized to podocytes and distal convoluted tubules (78). AR activity was detected in mesangial cells in culture (79), but not in vivo (77, 78). SDH mRNA expression was highest in the cortex, in Bowmans capsule, glomerular tuft, collecting duct, peritubular endothelial and interstitial cells, and moderate in

Downloaded from edrv.endojournals.org on April 19, 2006

976

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

jority of the published studies point toward activation of the polyol pathway and sorbitol accumulation in diabetes. In contrast to diabetic dogs showing nephromegaly, glomerular hypertrophy, mesangial expansion, BMT, and albuminuria, dogs fed galactose for 5 yr only developed thickening of the glomerular basement membrane (91). Kidney polyol accumulation was much more pronounced in galactose-fed dogs than in diabetic dogs, suggesting that the accumulation of polyol per se is not sufficient to produce diabetic glomerulopathy (91). Transgenic mice overexpressing human AR developed pathological changes in the kidney, i.e., thrombi of renal vessels and fibrinous deposits in Bowmans capsules (98). 5. Clinical evidence for a role of AR in diabetic kidney disease. Data about the polyol pathway and diabetic nephropathy in type 1 diabetic patients are rather scarce. In peripheral blood mononuclear cells obtained from type 1 diabetic patients with diabetic kidney disease, AR expression was increased compared with that in normal subjects, type 1 diabetics without nephropathy, and nondiabetic subjects with renal disease (99). Studies on AR gene polymorphisms have implicated the polyol pathway in the development of kidney complications, because the presence of the Z-2 allele or the T allele of the AR gene in type 1 diabetic subjects was associated with an increased risk for diabetic nephropathy (100, 101). Type 2 diabetic patients had higher serum and urine myo-inositol concentrations and sorbitol excretion than healthy controls (102, 103), but urinary fructose excretion was not different between the groups (103). Improvement of glycemic control reduced the urinary myo-inositol excretion (103). Serum myo-inositol concentrations were higher in patients with nephropathy than in those without nephropathy, but there were no differences in urinary myo-inositol concentrations (102). Renal AR expression was increased in type 2 diabetic patients compared with controls (77), especially in the glomerular mesangial area of patients with nephropathy (77). Reduced tubular SDH expression in diabetic patients was associated with interstitial fibrosis and thickened basement membranes (77). 6. Agents with effects on the AR system in diabetic kidney disease. Numerous experimental and clinical studies with different ARIs have implicated the diabetes-induced polyol pathway activation in the development of diabetic retinopathy and neuropathy, but only a few studies have investigated the influence of ARIs in the diabetic kidney. Despite the large number of compounds active against AR in vitro, only two classes of ARIs can be reported regarding in vivo activity: cyclic imides (mostly spirohydantoins) and carboxylic acid derivatives with sorbinil and tolrestat being the most representative members, respectively (104). However, due to toxicity or a lack of efficacy, sorbinil, tolrestat, and ponalrestat have been withdrawn from clinical trials. In the following will be focused mainly on epalrestat, the only ARI still available (104). a. ARIs. Tolrestat blocked the glucose-induced increases in sorbitol, fructose, and DAG levels, PKC activity, and angiotensinogen expression in proximal tubular cells (87). Epalrestat abolished the glucose-induced increases in TGF- and

PKC activity in cultured human mesangial cells, providing evidence for an interaction between these glucose-induced pathways (82). In type 1 diabetic rats, short-term oral administration of epalrestat prevented the increase in UAE and the reduction of anionic sites on the lamina rara externa of the glomerular basement membrane (105). In type 1 diabetic rats receiving epalrestat for 2 wk, renal cortical sorbitol concentration and the urinary enzyme excretion were not increased compared with untreated diabetic rats, suggesting that sorbitol accumulation leads to proximal tubular cell dysfunction and abnormal enzymuria (106). Long-term treatment with epalrestat ameliorated the decline of GFR and renal plasma flow (RPF) and mesangial expansion observed in placebo-treated diabetic rats, implicating the polyol pathway in functional and morphological changes in type 1 diabetes (107). In type 1 diabetes, 6 months of AR inhibition with ponalrestat or tolrestat reduced GFR in normo- and microalbuminuric patients, respectively (108, 109). However, 3 months of ponalrestat treatment had no effect on renal hemodynamics in patients with incipient nephropathy (110). In microalbuminuric type 2 diabetic patients, 5 yr treatment with epalrestat prevented the progression of incipient diabetic nephropathy (Fig. 2) (111). The development of new ARIs (112) may provide more potent and specific agents to block the polyol pathway, and future studies are warranted to elucidate their potential in the prevention of the development or progression of diabetic kidney disease. 7. Conclusion. In vitro data clearly demonstrate a link between the polyol pathway, AGEs, PKC, RAS, and TGF- activation in hyperglycemic conditions, which may stimulate the production of ECM proteins. Experimental studies implicate activation of the polyol pathway with sorbitol and fructose accumulation in short- and long-term diabetes, although results on myo-inositol levels and AR activity are not always consistent. Clinical data point toward increased AR activity and myo-inositol levels. Many ARIs have been withdrawn from clinical use due to disappointing results in the treatment of diabetic retinopathy and neuropathy. However, both experimental and clinical studies indicate beneficial effects of certain ARIs on diabetic structural and functional renal changes.

FIG. 2. Changes in UAE of microalbuminuric patients treated with (F) and without (E) epalrestat. Vertical bars indicate means SEM. *, P 0.01 vs. 0 yr. #, P 0.05 vs. patients without epalrestat. [Reproduced with permission from K. Iso et al.: J Diabetes Complications 15:241244, 2001 (111). Elsevier.]

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 977

III. Hemodynamic Factors A. Angiotensin II (Ang II)/renin-angiotensin system (RAS)

1. The Ang II system. Apart from the circulating RAS that regulates blood pressure, fluid, and electrolyte balance, many tissues, including the kidney, appear to have an independently regulated local RAS (113). Renin, produced in the juxtaglomerular cells of the kidney, converts angiotensinogen, derived primarily from hepatocytes, to inactive Ang I, which is then converted to biologically active Ang II by ACE (113). Ang II exerts hemodynamic and nonhemodynamic effects by binding to the Ang II type 1 (AT1) or Ang II type 2 (AT2) receptors. Most of the intrarenal actions of Ang II are mediated by AT1 receptors that signal through activation of phospholipases and PKC and inactivation of adenylate cyclase (113). The role of AT2 receptors in kidney has not yet been clearly defined, but involves the production of NO and prostaglandin F2 and signal transduction through activation of guanylate cyclase, phosphatases, and potassium channels (113). AT2 receptors might also be involved in mediating proliferation and apoptosis (114), and they counteract the effects of AT1 receptor-mediated Ang II actions. 2. Expression of Ang II/RAS in the normal kidney. All components of the RAS, i.e., renin, angiotensinogen, ACE, Ang II, AT1 and AT2 receptors are expressed in normal kidney (113). Renin mRNA was identified in cultured human mesangial cells and glomerular podocytes (115). In rat and human kidney, renin mRNA or protein expression was detected in glomerular endothelial cells and proximal and distal tubular cells (115, 116). Angiotensinogen mRNA was observed in cultured human glomerular podocytes and mesangial cells (115) and rat proximal tubule suspensions (116). In human kidney, a tubular angiotensinogen mRNA distribution was found (115). ACE mRNA was detected in cultured human mesangial cells and glomerular podocytes, in rat proximal tubules (116), and in tubules in human kidney (115). Most of the intrarenal Ang II is formed within the kidney (113). In rat kidney, AT1 receptors were detected in afferent arterioles, arcuate arteries, vasa recta, glomerular mesangial cells and podocytes, proximal tubules, thick ascending limb, collecting ducts, and medullary interstitial cells (117). AT2 receptors were localized in afferent arterioles, arcuate arteries, vasa recta, glomerular cells, proximal tubules, collecting ducts, and interstitial cells (117119). 3. In vitro evidence for Ang II on renal cells. High glucose stimulated the mRNA expression and protein synthesis of angiotensinogen in proximal tubular epithelial cells and involved activation of the polyol pathway and PKC (87). In cultured rat mesangial cells, high glucose increased Ang II production and AT2 receptor expression and reduced the levels of aminopeptidase A, a metalloprotease that degrades Ang II, which could contribute to the increase of Ang II (120, 121). In cultured mesangial cells, the high glucose-induced inhibition of collagenase activity, reduction of MMP-2 levels, and increase in TGF-1 secretion, resulting in decreased matrix degradation and increased matrix accumulation, are mediated by Ang II (121). Ang II-induced ET-1 production in glomerular mesangial cells is partially PKC-dependent (122)

and plays a role in the mitogenic effect of Ang II (123). Ang II induced connective tissue growth factor (CTGF) in cultured mesangial and tubular epithelial cells (124). Ang II induced apoptosis in cultured proximal tubular cells (125). 4. Experimental evidence for a role of Ang II in diabetic kidney disease. Although the role of RAS in diabetic nephropathy is indisputable, data concerning the influence of diabetes on systemic and intrarenal RAS have been conflicting (126). In experimental type 1 and type 2 diabetes, plasma levels of RAS components have generally shown suppression of the system, and the local glomerular RAS seems to be activated, but data are variable, which may be due to the degree of hyperglycemia or the time after onset of diabetes (126). STZdiabetic rats tended to have increased plasma and intrarenal levels of Ang II compared with control and insulin-treated rats (116). Studies investigating renin, angiotensinogen, and ACE mRNA in the proximal tubules (116) and glomeruli (119) in rats with STZ diabetes for 2 wk only showed an increase in renin mRNA in the proximal tubules. A downregulation of cortical and proximal tubular AT1 receptors (116) and of AT2 receptors in all kidney regions (119) was reported, suggesting that alterations in the balance of kidney AT1 and AT2 receptors may contribute to Ang II-mediated diabetic glomerular injury. In diabetic (mRen-2)27 transgenic rats, diabetic renal pathology was associated with intense renin mRNA and protein in proximal tubules and juxtaglomerular cells along with overexpression of TGF-1 and collagen IV mRNA in glomeruli and tubules, as well as a declining GFR and UAE (127). 5. Clinical evidence for a role of Ang in diabetic kidney disease. In type 1 diabetic patients, hyperglycemia was associated with increased plasma renin concentrations, whereas in patients with type 2 diabetes and nephropathy plasma renin levels were suppressed (126). However, measurements of circulating components of the RAS do not appear to accurately predict the state of activation of the local RAS (126). One study found stronger signals for renin, angiotensinogen, and ACE mRNA in mesangial and epithelial cells from hypertensive patients and patients with renal pathology, including some with diabetes (115). In renal biopsy specimens from type 2 diabetic patients, enhanced glomerular ACE staining was reported, especially in diabetics with glomerular nodular lesions (128). Another study reported reduced AT1 receptor mRNA levels in kidney biopsy samples from type 2 diabetics with nephropathy when compared with controls (129). Several studies investigating polymorphisms of the ACE and angiotensinogen genes that may predispose people with diabetes to nephropathy have yielded varying outcomes. The TT genotype of the angiotensinogen M235T polymorphism has been associated with diabetic nephropathy in type 1 diabetes (130), whereas other studies in type 1 and type 2 diabetic subjects failed to find an association (131133). The D-allele of the ACE insertion/deletion (I/D) polymorphism has been identified as a risk factor for increased progression of diabetic glomerulopathy in microalbuminuric type 1 diabetic patients (134). However, other studies in type 1 (132) and type 2 diabetic patients (133) did not support a role for ACE I/D polymorphism in the development of di-

Downloaded from edrv.endojournals.org on April 19, 2006

978

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

abetic nephropathy. A metaanalysis of the ACE I/D polymorphism and risk of diabetic nephropathy, including 11 studies in type 1 and 10 studies in type 2 diabetic patients, suggested that the ACE I/D polymorphism contributes to the genetic susceptibility to diabetic nephropathy in Japanese but not in Caucasian type 2 diabetic patients (135). In Caucasian type 1 diabetic patients, comparison of data was complicated by differences between study populations, but a trend toward a protective effect of the II genotype on the development of increased UAE was observed (135). The risk for diabetic nephropathy in type 1 diabetic patients was similar in carriers and noncarriers of the AT1 receptor A1166C polymorphism (132). 6. Agents with effects on the Ang II system in diabetic kidney disease. The best known agents to target the RAS are ACEi and the newer ARBs. ACEi act in part by reducing plasma Ang II levels and increasing plasma bradykinin levels (136). Bradykinin accumulation has been associated with side effects, including cough and angioedema, but may also contribute to the antihypertensive and vasculoprotective effects of ACEi. ACEi do not provide a complete inhibition of Ang II activity, because Ang II can be formed through non-ACE pathways. ARBs might exert a more specific and complete blockade of the RAS through their inhibition of the actions of Ang II at the AT1 receptor, but this results in higher circulating Ang II levels (137). The biological activity of Ang II or its metabolites may be more directed to other Ang II binding sites with potential adverse effects (138). In addition, ARBs raise bradykinin levels through stimulation of AT2 receptors by elevated Ang II levels. Thus, ACEi and ARBs potentially have additive effects on both the RAS and bradykinin levels (136). The potential role of oral renin inhibitors that decrease Ang II levels in humans and of AS-ODNs designed to target renin, angiotensinogen, ACE, or AT1 receptors in the therapy of diabetic nephropathy remains to be investigated. a. ACEi. ACEi consistently limited progressive renal injury in experimental models of STZ-induced diabetes (139). In Wistar fatty rats, an experimental model of type 2 diabetes with progressive kidney disease, enalapril ameliorated both existing proteinuria and the progression of proteinuria and preserved glomerular structure (5). In microalbuminuric type 1 diabetic subjects, perindopril reduced glomerular BMT and tended to reduce interstitial fibrosis (140), and enalapril prevented glomerular growth (141) and progression of glomerulopathy, indicating that ACE inhibition may influence renal structural changes (142). However, in a small cohort of normotensive type 1 diabetic patients with albuminuria and diabetic glomerulopathy, enalapril treatment did not affect renal structure (143). In normotensive normoalbuminuric type 1 diabetic patients, acute ACE inhibition by enalapril caused a decline in filtration fraction and in blood pressure but had no effect on UAE (144); 6 wk of captopril treatment had no effect on GFR and UAE (145), but treatment with enalapril for 3 months reduced UAE without effect on GFR or RPF (144). In normotensive microalbuminuric type 1 diabetic patients, UAE and blood pressure were reduced, whereas GFR was unaffected by long-term treatment with ramipril (146) or perindopril (147). In micro- and

macroalbuminuric type 1 diabetic patients, acute ACE inhibition by enalapril did not change GFR, but it reduced blood pressure, UAE, filtration fraction, and renal vascular resistance, whereas RPF tended to rise (148). Six months of enalapril treatment induced similar changes except for GFR and RPF; GFR was reduced, whereas RPF remained unchanged (149). Enalapril was more effective in reducing UAE in proteinuric type 1 diabetic patients than antihypertensive patients (148). In normotensive type 1 diabetics with nephropathy treated for 8 yr with captopril, systemic blood pressure and albuminuria remained unchanged and only a minimal loss of GFR was seen when compared with patients who were not treated with captopril (150). However, captopril treatment was associated with a 50% risk reduction of the combined end points of death, dialysis, and transplantation (Fig. 3) (151). In macroalbuminuric type 1 diabetic patients with mild to moderate hypertension, treatment with enalapril reduced UAE, total protein excretion, and blood pressure, but had no effect on GFR and RPF (152). In microalbuminuric type 2 diabetic subjects, perindopril also reduced glomerular BMT and tended to reduce interstitial fibrosis (140). In micro- and macroalbuminuric type 2 diabetic patients with urinary podocyte excretion, trandolapril treatment reduced the number of podocytes excreted in the urine (153). The abnormalities in size-selective function of the glomerular barrier in type 2 diabetics with overt nephropathy were not ameliorated by ACEi or calcium channel blockade at variance to type 1 diabetes (154). Enalapril attenuated the decline in renal function and reduced the extent of albuminuria in normotensive, normo- and microalbuminuric patients with type 2 diabetes (155, 156). In hypertensive type 2 diabetic patients with normo-, micro-, or macroalbuminuria, treatment with enalapril was associated with a greater reduction in UAE than with nifedipine in the entire patient group, and especially in those with microalbuminuria. In the macroalbuminuric patients, the rate of deterioration in renal function was also attenuated by treatment with enalapril (157). b. ARBs. In diabetic Wistar fatty rats, candesartan treatment for 4 wk prevented an increase in proteinuria and development of glomerular lesions (5). In Zucker diabetic fatty (ZDF) rats, a model of obese type 2 diabetes, 19 wk of treatment with olmesartan suppressed proteinuria, corrected hyperlipidemia and hypoalbuminemia, and reduced tubular and glomerular damage in ZDF rats, indicating that the renoprotective effect of olmesartan is, at least in part, independent of its antihypertensive effect (158). Recent clinical trials with irbesartan in type 2 diabetic patients with microalbuminuria (IRMA2) (Fig. 4) (159) or nephropathy (IDNT) (160) and with losartan in patients with type 2 diabetes and nephropathy (RENAAL) (161) have demonstrated the renoprotective effects of ARBs independently of their blood pressure-lowering effect. A clinical trial with valsartan in high-risk patients with type 2 diabetes is ongoing (ABCD-2C) and will compare the effect of intensive vs. moderate control of blood pressure on diabetic complications and mortality. c. Combination of ACEi and ARBs. In renin transgenic rats, the combination of enalapril and losartan resulted in higher

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 979

athy and hypertension despite ACEi treatment, the addition of irbesartan or candesartan, respectively, reduced blood pressure and albuminuria more effectively than ACEi alone (165, 166). In the Candesartan and Lisinopril Microalbuminuria Study (CALM) in microalbuminuric type 2 diabetic patients with hypertension, combination therapy afforded greater reductions in blood pressure and albuminuria than either drug alone (167). 7. Conclusion. Data regarding the renal expression of RAS components are inconsistent. Furthermore, the contribution of ACE or angiotensinogen polymorphisms to the development or progression of diabetic nephropathy remains controversial. Nevertheless, the beneficial effects of ACEi and/or ARBs on diabetic renal structural and functional changes beyond their blood pressure-lowering effect provides substantial experimental and clinical evidence for a deleterious role of RAS components in diabetic nephropathy. The combination of ACEi and ARBs seems to have an additive effect, at least on blood pressure and UAE. The effects of ACEi on renal structure and function may be partly explained by effects on growth factors and cytokines, metabolic pathways, and signaling molecules.
B. Endothelin (ET)

FIG. 3. Cumulative incidence of events in patients with diabetic nephropathy in the captopril and placebo groups. A, The cumulative percentage of patients with the primary end point: a doubling of the baseline serum creatinine concentration to at least 2.0 mg/dl. B, The cumulative percentage of patients who died or required dialysis or renal transplantation. The numbers at the bottom of each panel are the numbers of patients in each group at risk for the event at baseline and after each 6-month period. [Reproduced with permission from E. J. Lewis et al.: N Engl J Med 329:1456 1462, 1993 (151). Massachusetts Medical Society.]

1. The ET system. The ET system comprises the ETs, two receptors, and two activating peptidases. The ETs are a family of structurally and functionally related peptides, more in particular ET-1, the most potent vasoconstrictor known, ET-2, and ET-3. Preproendothelins, the ET precursors, are cleaved by endopeptidases to form inactive intermediates termed big ETs. Big ETs are then cleaved by ET-converting enzymes (ECEs) to form the final products (168, 169). In mammals, the two ET receptors, ETA and ETB, signal through the activation of G-proteins, phospholipase C, and PKC. ETA receptors are involved in vasoconstriction and cell proliferation, whereas ETB receptors binding all ETs mediate NO release and transient vasodilation (168, 169). ETs are normally not circulating hormones but act as paracrine and autocrine factors. The (patho)physiological roles of ETs in various organs have been described in detail (170). In the kidney, the ET system regulates renal hemodynamics, water and sodium homeostasis, cell proliferation, and matrix formation (168 170). 2. Expression of ET in the normal kidney. ET-1, ET-2, and ET-3, ECE-1, and receptors ETA and ETB are present in the kidney (169, 171). ET-1 is found in all parts of the nephron. ET-1 is synthesized and secreted by cultured glomerular mesangial (122, 172174), epithelial (175), and endothelial cells (176) and proximal tubular cells (177). In porcine kidney, the concentration of immunoreactive ET-1 was highest in renal inner medulla and very low in cortex (178). In rat kidney, ET-1 mRNA was demonstrated in cortical and medullary collecting ducts (179). In human kidney, mature ET and big ET-1 localized to the endothelium of glomeruli and intrarenal blood vessels (176). All three ET isoforms were detected by RT-PCR in human homogenates of renal medulla, cortex, and vessels (176). ET-3 is distributed differently along the nephron than ET-1 in rat kidney (180). ECE-1 localized to

plasma renin levels than either drug alone, suggesting an additive inhibition of the RAS by combination therapy (162). In an experimental model of diabetic nephropathy, combination therapy with valsartan and perindopril lowered blood pressure and improved severe diabetic glomerulosclerosis better than monotherapy but was equally effective at ameliorating renal function and reducing albuminuria (163). The combination of irbesartan and captopril in diabetic spontaneously hypertensive rats demonstrated additive hypotensive and antialbuminuric effects compared with monotherapy (164). These data, however, do not allow clear distinction between the additive effects of combination therapy on blood pressure vs. additive intrinsic renoprotective effects. In type 1 and type 2 diabetic patients with nephrop-

Downloaded from edrv.endojournals.org on April 19, 2006

980

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

FIG. 4. Incidence of progression to diabetic nephropathy during treatment with 150 mg of irbesartan daily, 300 mg of irbesartan daily, or placebo in hypertensive patients with type 2 diabetes and persistent microalbuminuria. The difference between the placebo group and the 150-mg group was not significant (P 0.08 by the log-rank test), but the difference between the placebo group and the 300-mg group was significant (P 0.001 by the log-rank test). [Reproduced with permission from H.-H. Parving et al.: N Engl J Med 345:870 878, 2001 (159). Massachusetts Medical Society.]

vascular endothelial and tubular epithelial cells in the cortex and medulla of human kidney (171). Both ETA and ETB receptor mRNAs were expressed in human mesangial cells (181), in rat glomeruli (182), and in normal human renal cortex and medulla with ETB predominating (183). 3. In vitro evidence for ET effects on renal cells. Various agents increase ET-1 production by mesangial cells, including hyperglycemia, PDGF, TGF-1, thrombin, Ang II, and ET-1 (181). ET-1 release is also increased by thrombin and bradykinin in glomerular endothelial cells (184) and by TGF- in collecting duct cells (185). Other factors such as shear-stress due to glomerular hyperfiltration (186) and urine flow (187) have been shown to stimulate ET-1 synthesis or release. Furthermore, AGEs induced ET-1 mRNA expression in cultured proximal tubular cells (188). High glucose modified the responses of mesangial cells to ET-1 (181). For example, hyperglycemia augmented ET-1-stimulated 1 (IV) collagen production and MAPK activity in mesangial cells, effects that are PKC dependent and associated with altered translocation of PKC and PKC (181, 189). ET-1 stimulates mesangial cell proliferation, contraction, and ECM accumulation (176). 4. Experimental evidence for a role of ET in diabetic kidney disease. Studies examining systemic and intrarenal ET-1 levels in diabetic animals have yielded conflicting results. In STZdiabetic rats, plasma ET-1 levels have been undetectable (190), unchanged (191), enhanced (187, 192), or suppressed (193) compared with control rats. Urinary ET-1 excretion was increased in STZ-diabetic rats, in parallel with enhanced proteinuria (139). Renal ET-1 levels were reported to be unchanged (190) or reduced (191) and glomerular ET-1 levels were enhanced (182) in STZ-diabetic vs. control rats. In early STZ-diabetic rats, no difference in glomerular ETA receptor characteristics has been found; however, a reduction in ETB density, as well as a reduction in glomerular Ang II receptor density has been reported (194). The mRNA levels for ETA and ETB did not change in STZ-diabetic rats (182). In type 2 diabetic rats, glomerular and tubulointerstitial ET-1 mRNA and protein expression was higher than in nondiabetic rats (195). The intrarenal ET system could be affected indepen-

dently of the systemic ET-1 system. Discrepancies may be related to the degree of hyperglycemia, the renal localization, or varying duration of diabetes. Up-regulation of the renal ET system might favor the development of renal lesions, as suggested by in vivo studies in transgenic mice and rats. ET-1 transgenic mice with only slightly elevated tissue and plasma ET-1 concentrations developed glomerulosclerosis, interstitial fibrosis, renal cysts, and a progressive decline in GFR (196). ET-2 transgenic rats with a high renal transgene expression almost exclusively within glomeruli developed glomerulosclerosis and albuminuria without a change in GFR (197). 5. Clinical evidence for a role of ET in diabetic kidney disease. Few clinical studies have been published on ET in diabetic kidney disease. In patients with uncomplicated type 1 diabetes, the plasma concentrations of ET-1 were increased and directly correlated to those of von Willebrand factor, and inversely correlated to plasma concentrations of fibronectin (198). Plasma ET-1 levels were higher in nonsmoking normotensive type 2 diabetic patients than in controls and higher in microalbuminuric vs. normoalbuminuric patients (199). Plasma ET-1 concentrations were increased in hypertensive type 2 diabetic patients as compared with healthy control subjects but were not different from those in normotensive patients (200). In contrast, plasma ET-1 levels were equally elevated in type 1 and type 2 diabetic patients and hypertensive nondiabetic patients compared with healthy controls, but the diabetic groups included patients with and without hypertension (201). Plasma ET-1 levels did not correlate with clinical parameters of diabetic disease progression in diabetic patients, but correlated with age and systolic blood pressure in healthy controls (201). In addition, plasma and urinary ET-1-like immunoreactivity values correlated positively with the severity of diabetic nephropathy in type 2 diabetics (202). In contrast, another study found no differences in the diurnal urinary excretion of immunoreactive ET among diabetic patients and patients with nondiabetic diseases (203). 6. Agents with effects on ET in diabetic kidney disease. Several agents are available to inhibit the action of ET, including

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 981

neutralizing antibodies against ET and ET receptors and AS-ODNs to preproendothelin-1, ECE, and the ETA receptor. However, the potential use of these substances in the treatment of diabetic renal changes has not been explored, except for some in vitro studies. Ample experimental studies focused on the renoprotective effects of ET receptor antagonists. The ET system can be targeted indirectly by ACEi and PKC inhibitors. a. Neutralizing antibodies. Addition of a neutralizing anti-ET antibody to mesangial cell cultures markedly augmented the secretion and activation of MMP-2, whereas addition of exogenous ET-1 inhibited the synthesis of MMP-2. These results implicate ET as a potential factor in pathophysiological matrix turnover in the glomerulus (204). b. AS-ODNs. AS-ODNs targeting preproendothelin-1 mRNA were delivered into cultured human mesangial cells and inhibited ET-1 secretion and the cell proliferation. These results identify ET-1 as one of the autocrine growth factors of human mesangial cells that may be important in pathophysiological conditions characterized by mesangial proliferation (205).

c. ET receptor antagonists. The effects of ETs can be blocked by the administration of nonselective ET receptor antagonists (bosentan, PD 142893, TAK-044, LU 224332), selective ETA antagonists (FR139317, BQ-123, BMS-193884, LU 135252, PD156707, and EMD 94246), or ETB antagonists (RES-701-1, BQ-788). PD 142893 reduced UAE in diabetic animals (206). In nondiabetic and STZ-diabetic (mRen-2)27 rats, oral administration of bosentan for 12 wk normalized systolic blood pressure and attenuated the diabetes-associated decline in GFR (127). Despite producing normotension, severe diabetic renal pathology was not prevented by bosentan, suggesting dissociation of ET, UAE, and hypertension from the structural injury in this diabetic model (Fig. 5) (127). Selective ETA blockade with FR139317 has shown protective effects in experimental diabetic glomerulopathy (192). In STZ-diabetic rats, mRNA levels for various collagens, laminin, and growth factors were elevated (192) and glomerular proliferating cell nuclear antigen, c-myc, c-fos, and c-jun mRNA levels increased with progression of diabetic nephropathy (207). These changes were reduced by FR139317 (207), suggesting that ET may play a role in ECM production. Thus, ET inhibition may protect against diabetic glomerular injury, pos-

FIG. 5. Histopathology of the kidney cortex in nondiabetic and diabetic transgenic (mREN-2)27 rats treated for 12 wk with either valsartan or bosentan. Sections are stained with periodic acid-Schiff. G, Glomerulus. A, In nondiabetic rats, there is minimal evidence of glomerulosclerosis. B, Diabetic rats have severe glomerulosclerosis, arteriolar hyalinization (asterisk). Valsartantreated nondiabetic (C) and diabetic rats (D) and bosentan-treated nondiabetic rats (E) have minimal evidence of glomerulosclerosis. Bosentan-treated diabetic rats (F) have severe glomerulosclerosis and cortical interstitial fibrosis (arrow; magnification, 300). [Reproduced with permission from D. J. Kelly et al.: Kidney Int 57:18821894, 2000 (127). Blackwell Publishing.]

Downloaded from edrv.endojournals.org on April 19, 2006

982

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

sibly by interference with growth factors and growth-related genes (192). LU 135252 prevented the Ang II-induced increase in tissue ET-1 content and functional ECE activity in rats (208). Furthermore, LU 135252 prevented renal histological alterations in STZ-diabetic rats, and decreased urinary ET-1 excretion, but had no effect on UAE (209). Both LU 135252 and LU 224332 reduced proteinuria and completely normalized the renal matrix expression of type IV collagen and fibronectin in hyperglycemic rats with STZ-induced diabetes (210). d. ACEi. Lisinopril attenuated the increased renal ET-1 expression in STZ-diabetic rats and had better blood pressure-lowering but similar renoprotective effects than the unselective ETA/ETB receptor antagonist, PD 142,893 (206). Treatment with enalapril or nicardipine similarly reduced the elevated plasma ET-1 levels in hypertensive type 2 diabetic patients to control levels. The change in plasma ET-1 level was not correlated to the change in blood pressure or to UAE (200). e. PKC inhibition. The ET-1-induced MAPK activation in mesangial cells is PKC dependent and associated with altered translocation of PKC and PKC (189). Infusion of the specific PKC inhibitor 1-(6-isoquinolinesulfonyl) piperazine reversed the down-regulation of ET receptors in association with normalization of PKC activity in STZ-diabetic rats (194). 7. Conclusion. Various renal cell types can secrete ET-1 in vitro, which can be stimulated by glucose, Ang II, TGF-, and PDGF. Experimental data regarding plasma and renal ET-1 levels are conflicting, but experiments in transgenic animals clearly show that up-regulation of ET-1 or ET-2 may favor the development of structural and functional renal changes. In type 1 and type 2 diabetic patients, increased plasma ET-1 levels correlate with the severity of diabetic nephropathy. In vitro application of neutralizing ET antibodies and AS-ODNs indicates ET-1 as a potential factor in mesangial proliferation and matrix turnover. A strong argument in favor of ET-1 as a mediator of renal injury derives from studies with ET receptor antagonists in experimental diabetes. Independent of their blood pressure-lowering effect, ET receptor antagonists reduced renal ET-1 content, urinary ET-1 excretion, and the production of ECM proteins; lowered UAE; and reduced renal expression of TGF- and PDGF-B.
C. Nitric oxide (NO)

structural integrity (213). Endogenous inhibitors of NOS are NG-monomethyl-l-arginine (l-NMMA) and asymmetric dimethylarginine (ADMA), substances naturally occurring in human plasma (214). ADMA is mainly cleared by the enzyme dimethylarginine-dimethylaminohydrolase (DDAH) and partly by renal secretion (215). 2. Expression of the NO system in the normal kidney. All three NOS isoforms are present in the kidney (211). Their localization and that of alternative splice variants have been described in detail (216). The renal medulla is the main site for NO synthesis in the kidney (216). NOS1 mRNA and protein were detected abundantly in the macula densa and in some thick ascending limb cells in rat and human kidney (217, 218). In addition, NOS1 immunoreactivity has been demonstrated in the endothelium of glomerular efferent arterioles, inner medullary collecting ducts, and the parietal epithelium of Bowmans capsule in rat kidney (217, 218). In rat and human kidney, NOS3 was typically detected in the endothelium of preglomerular, glomerular, and postglomerular vessels, whereas NOS2 was hardly detected (217, 219). In human glomeruli NOS3 mRNA was constitutively expressed, whereas NOS2 mRNA expression was barely found, and NOS1 mRNA was not detectable (220). 3. In vitro evidence for NO effects on renal cells. Most studies in cultured renal cells have provided evidence that diabetes is a state of NO deficiency (211). For example, high glucose attenuated the detectable NO, blunted the NO responses to NOS3 agonists in human endothelial cells (221), and inhibited NO synthesis in murine mesangial cells (222). Possible mechanisms whereby high glucose decreases NO bioavailibility include NO capture by glucose (221), superoxide anion generation (223), endothelial ADMA accumulation (215), l-arginine depletion, and reduced tetrahydrobiopterin stability and availability (222). Other studies, however, in cultured mesangial cells, documented the stimulation of NO production in hyperglycemic conditions (224, 225), which may involve NOS2, PKC, and AR activation (224). NO deficiency as well as increased NO production may promote ECM accumulation in the mesangium (224, 226). The in vitro assessment of renal vascular reactivity in isolated rat kidneys and isolated renal arteries has provided more controversial evidence, suggesting increased, normal, or decreased NO synthesis and activity. Some of the discrepancies between different studies could relate to the method of preconstriction of isolated vessels (211). 4. Experimental evidence for a role of NO in diabetic kidney disease. Research on the effect of experimental diabetes on the renal expression of NOS isoforms has also yielded conflicting results (reviewed extensively in Ref. 211). At 7 d of STZdiabetes, total cortical NOS activity was reduced and associated with decreased NOS1 mRNA and NADPH diaphorase staining in the macula densa, whereas cortical NOS3 was not different between diabetic and control rats (227). In contrast, after 2 wk of STZ-diabetes, NOS3 protein levels and immunostaining were elevated in glomerular endothelial cells and preglomerular vessels in diabetic vs. control rats (228, 229). However, another study found no differences in renal cortical expression of NOS isoforms or in diaphorase staining

1. The NO system. NO is a widespread signaling molecule that plays a major role in nearly every cellular and organ function in the body. The enzymatic formation of NO from l-arginine, generating l-citrulline as a coproduct, is catalyzed by NOS. Three mammalian NOS isoforms have been identified, neuronal (nNOS), endothelial (eNOS), and macrophage or inducible NOS (iNOS), more recently termed NOS1, NOS2, and NOS3, respectively (211). Their structure, function, and inhibition has been reviewed extensively (212). NO mediates its effects through activation of guanylate cyclase, resulting in increased levels of cyclic GMP (cGMP). In the kidney, NO is involved in the regulation of RPF, GFR, sodium excretion, extracellular fluid volume, and the maintenance of renal

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 983

between control and diabetic rats (230). After 4 wk of STZdiabetes, NOS1 immunostaining was modestly enhanced in macula densa (218), whole kidney NOS2 and NOS3 mRNA levels were unchanged (231), but NOS3 protein expression was increased, and NOS1 protein expression was unaltered in whole kidney samples (232). After 6 wk of STZ-diabetes, medullary NOS1 and NOS3 mRNA levels were elevated, immunostaining for NOS1 and NOS3 was increased in the proximal straight tubules and medullary thick ascending limb (233), and enhanced NOS3 immunostaining was also found in purified renal vascular trees (Fig. 6) (234). Data concerning the role of NO in glomerular hyperfiltration in experimental diabetes are also conflicting (reviewed extensively in Ref. 211). The majority of studies in type 1 diabetic rats found glomerular hyperfiltration in the presence of increased urinary NO2 and NO3 (NOx) excretion, supporting the notion that increased NO synthesis contributes to diabetic hyperfiltration (228, 233238). The NO-donor glyceryl trinitrate induced renal vasodilation in control but not in diabetic rats, providing further evidence for a role of enhanced NO production and/or signaling that is not changed by additional NO (236). However, other studies suggested a defect in renal NO production and/or action in experimental diabetes. In rats with type 1 diabetes for 1 to 2 wk, increased CrCl and renal hypertrophy were accompanied by a decreased NOx excretion and unchanged plasma NO metabolites when compared with nondiabetic controls (227, 230). Reduced NO-dependent renal vasodilation (239, 240) and decreased glomerular NO-dependent cGMP generation (239, 241) were reported in diabetic rats compared with controls. The impaired glomerular NO-dependent cGMP generation may be mediated by thromboxane A2 and PKC activation (241) or by an impaired activity of guanylate cyclase (231). NO has been generally considered as the principal mediator of endothelium-dependent vasodilation. En-

dothelium-dependent (i.e., NOS3-dependent) vasodilation is impaired in diabetic animal models and in humans with type 1 and 2 diabetes (242). In the renal circulation of STZ-diabetic rats in vivo, the vasodilation to intrarenal acetylcholine was reduced compared with control rats, suggesting impaired endothelium-dependent vasodilation (243). NO-dependent vasodilation was enhanced, whereas the vasodilation mediated by endothelium-derived hyperpolarizing factor was severely impaired (243). 5. Clinical evidence for a role of NO in diabetic kidney disease. Normoalbuminuric type 1 diabetic subjects had a higher whole-body NO synthesis than control subjects as quantified by measuring the urinary excretion of 15N-nitrate after the iv administration of l-[15N]2-arginine (244). A weak positive relationship was found in the diabetic subjects between the amount of 15N-nitrate excreted and CrCl, supporting an association between NO production and hyperfiltration (244). In microalbuminuric type 1 diabetic patients, serum NOx was independently associated with both UAE and GFR, suggesting a strong link between NO, glomerular hyperfiltration, and microalbuminuria (245). In type 2 diabetic subjects, plasma levels of nitrite, but not nitrate, were higher than in nondiabetic subjects, increased with the progression of diabetic nephropathy, and were positively correlated with AGEs (246). In contrast, one study found serum NOx levels to be lower in type 2 diabetic patients than in controls, and to be higher in males than in females (247). Urinary NOx excretion was higher in normo- and microalbuminuric type 2 diabetic patients with glomerular hyperfiltration compared with controls and macroalbuminuric patients without glomerular hyperfiltration, providing evidence that NO may also contribute to the glomerular hyperfiltration in type 2 diabetes (248). Improvement of metabolic control did not change plasma or urinary NOx levels (249). Plasma ADMA

FIG. 6. Immunocytochemical staining for eNOS in control (left) and diabetic (right) kidneys. Staining is detectable in glomerular capillaries and in afferent and efferent arterioles, visible at the vascular pole of the glomeruli. More capillary profiles are stained in the diabetic glomerulus. [Reproduced with permission from A. S. De Vriese et al.: Kidney Int 60:202210, 2001 (234). Blackwell Publishing.]

Downloaded from edrv.endojournals.org on April 19, 2006

984

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

levels correlated with plasma glucose in diabetic and nondiabetic subjects (250). Circulating ADMA levels were higher in untreated type 2 diabetic patients than in matched controls (251, 252), and improvement of glycemic control was associated with decreased plasma ADMA levels (253). Recently, increased ACE activity has been implicated in the mechanism of increased ADMA levels in type 2 diabetic patients (252). However, it is not known whether increased ADMA levels play a role in the development or progression of diabetic nephropathy. 6. Agents with effects on the NO system in diabetic kidney disease. The most frequently used pharmacological inhibitors of NOS are the nonselective l-arginine analogs including Ng-nitrol-arginine methyl ester (l-NAME), l-NMMA, N--nitro-larginine (l-NNA), and aminoguanidine (212). S-Methyl-lthiocitrulline is a partially selective NOS1 inhibitor, and l-imino-ethyl-lysine is a partially selective inhibitor of NOS2 (212). ACEi and ARB, inhibitors of AGE formation and antioxidants, also influence the NO system. a. l-Arginine analogs. Acute systemic infusion of NOS blockers decreased GFR and RPF more in diabetic rats than in control rats (228, 236, 238, 254). Administration of l-NAME to the kidney caused a more pronounced renal vasoconstriction in diabetic rats than in control rats, suggesting that the basal vasodilation in the diabetic renal microcirculation can be attributed to increased NO generation (234). Chronic treatment with l-NAME in STZ-diabetic rats reduced GFR (229, 231, 254, 255) and urinary NOx excretion (229, 231, 255), attenuated kidney and glomerular growth (229, 231), and reduced the afferent arteriolar dilation and NOS3 fluorescence intensity in arterioles and glomeruli, providing evidence that enhanced NO synthesis by NOS3 in afferent arteriolar and glomerular endothelial cells contributes to diabetic glomerular hypertrophy and hyperfiltration (229). However, other studies provided discrepant results. Renal vasoconstriction and decreases in GFR and RPF in response to l-NAME were similar in diabetic and control animals (235, 256). Finally, a blunted vasoconstriction in response to systemic NOS blockade with l-NMMA (240) and l-NNA (257) was observed in diabetic rats as compared with controls, suggesting a decreased NO-mediated influence. The response of GFR and filtration fraction to l-NMMA was impaired in type 2 diabetic patients compared with controls, indicating that the regulation of glomerular hemodynamics by NO is altered in type 2 diabetes (258). Treatment of STZdiabetic rats for 6 months with a dose of l-NAME that did not raise blood pressure had no effect on GFR, urinary albumin, or NOx excretion (35). More selective NOS inhibitors allowed to elucidate the contribution of individual NOS isoforms in the pathogenesis of diabetic hyperfiltration. NOS1 inhibition with S-methyl-l-thiocitrulline nearly normalized GFR in hyperfiltering diabetic rats (218). Administration of l-imino-ethyl-lysine did not affect blood pressure, GFR, RPF (228), urinary NOx excretion, or kidney weight in diabetic rats (231). These results indicate that NOS1 contributes to altered renal NO production and hemodynamics but do not support a role for increased glomerular activity of NOS2 in experimental diabetes (228, 231).

b. ACEi/ARBs. Two weeks of treatment with quinapril or candesartan normalized UAE, renal NADPH oxidase immunostaining, plasma lipid peroxidation, kidney hydrogen peroxide production in early STZ-diabetes in rats, renal endothelial NOS3 expression, and renal nitrotyrosine expression, suggesting that the increased expression of NADPH oxidase and NOS3, leading to increased nitrooxidative stress, could be involved in the pathology of diabetic kidney disease (232). Imidapril and L-158,809 equally ameliorated the renal up-regulation of lipopolysaccharide-stimulated NOS2 expression of STZ-diabetic rats, suggesting that Ang II activation may play a role in enhanced NOS2 expression in diabetic nephropathy (259). Treatment with perindopril decreased serum ADMA levels in a small number of type 2 diabetic patients (252), suggesting that increased ACE activity may be involved in the endothelial dysfunction associated with ADMA elevation present in patients with noncomplicated type 2 diabetes. c. Antioxidants. In vitro, the effects of high glucose on DDAH activity and ADMA concentrations were reversed by superoxide dismutase (SOD), suggesting that oxidative stress plays a role in the high glucose-induced impairment of DDAH activity (215). The impaired responsiveness of afferent and efferent arterioles from STZ-diabetic rats to l-NNA was reversed by exogenous SOD, indicating that suppressed SOD activity reduces the tonic influence of NO on renal arterioles during the early stage of experimental diabetes (257). d. Inhibitors of AGE formation. One study compared the effects of aminoguanidine with two other inhibitors of NOS, l-NAME and methylguanidine, on the development of experimental diabetic kidney disease (35). Aminoguanidine prevented increases in UAE, urinary NOx, and renal AGE levels, whereas l-NAME and methylguanidine did not, suggesting that the beneficial effects of aminoguanidine may be mediated by decreased AGE formation rather than by NOS inhibition (see also Section II.A). Long-term treatment with aminoguanidine reduced TNF- and NOS2 expression, intraglomerular NOx production, and proteinuria in STZdiabetic rats, providing evidence that the AGE-cytokine-NO sequence pathway could be an important mechanism in the development of diabetic nephropathy (260). e. Anti-VEGF antibodies. Inhibition of VEGF decreased hyperfiltration, glomerular hypertrophy and UAE, and prevented the up-regulation of NOS3 expression in glomerular capillary endothelial cells of STZ-diabetic rats, indicating that NO may be a downstream mediator of VEGF in the kidney (261). 7. Conclusion. In vitro data regarding the NO system are conflicting. Hyperglycemia-induced NO production as well as NO deficiency contribute to ECM accumulation in cell cultures. Renal NOS isoform expression in early experimental diabetes varies considerably between different studies, although the majority reports increased NOS3 expression. In vivo hemodynamic studies that determined GFR and RPF by clearance techniques suggested increased renal NO production/activity in hyperfiltering diabetic rats, whereas studies reporting the contrary used CrCl or other techniques (211).

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 985

Discrepancies may also relate to the absence or presence of insulin treatment. Furthermore, urinary NOx measurements are only an indirect indicator of renal NOS activity. Clinical data support a role for NO in glomerular hyperfiltration in both type 1 and type 2 diabetes. The role of elevated circulating ADMA levels in the pathogenesis of diabetic nephropathy has not been elucidated. Inhibition of NOS and NOS1 by nonselective or more selective l-arginine analogs reduces GFR, giving further support that NOS1 contributes to altered glomerular hemodynamics in diabetes. The effect of NOS blockers on renal vascular resistance is highly discrepant between different studies.

IV. Intracellular Factors A. Diacylglycerol (DAG)-protein kinase C (PKC) pathway

(272). The concomitant increase in total cellular DAG levels (272275) suggests that glucose-induced increases in DAG may contribute to the activation of glomerular PKC observed in early diabetes. In cultured mesangial cells or diabetic glomeruli, hyperglycemia-induced PKC activation has been linked to several abnormalities, i.e., increased arachidonic acid release and production of prostaglandins, increased expression of fibronectin and type 1 (IV) collagen, decreased NaK-ATPase activity (272), stimulation of ERK (273), nuclear factor-B (NF-B) dependent proliferation (276), and suppression of MAPK phospatase-1 (277). PDGF induced de novo synthesis of PKCII and activation of PKC-, -, -, and - in cultured mesangial cells (278). Thus, cytokines can induce PKC activation, but PKC activation also stimulates the expression of cytokines such as VEGF (279) and TGF-1 (272). Overexpression of glucose transporter 1 also led to activation of PKC (81). 4. Experimental evidence for a role of DAG/PKC in diabetic kidney disease. PKC activity was increased in the glomeruli of type 1 diabetic rats from 2 wk after the onset of diabetes (194, 274) until 24 wk of diabetes (52, 272, 280, 281). Concomitant increases in glomerular DAG content have been reported (52, 282). More particularly, PKC was activated in the glomeruli of short-term STZ-diabetic rats as assessed by a change in the subcellular distribution of PKC specific activity (274). However, total PKC activity was not different between glomeruli from control and diabetic rats (274). Twelve weeks of STZ diabetes was associated with activation of the PKC and -I isoforms, as demonstrated by their increased protein expression in the membranous fraction and increased phosphorylation in glomeruli of diabetic rats (272). Furthermore, glomerular PKC activity was also increased in db/db mice compared with nondiabetic control animals (282). PKC activation has been reported to mediate the down-regulation of glomerular low-affinity ET-1 receptors (194). Similar to in vitro studies, PKC activation may contribute to early renal dysfunction in experimental diabetes by the alteration of prostaglandin production and NaK-ATPase activity and the overexpression of TGF-1 and ECM components (272). 5. Clinical evidence for a role of PKC in diabetic kidney disease. In skin fibroblasts cultured in normal and high glucose concentrations, total PKC activity and DAG content were higher in cells from type 1 patients with nephropathy than patients without or control subjects. Recently, a population- and family-based study demonstrated that type 1 diabetic carriers of the T allele of the 1504C/T single nucleotide polymorphism (SNP) or the G allele of the 546C/G SNP in the promoter of PKC-1 gene have an increased risk for diabetic nephropathy compared with type 1 diabetic noncarriers (283). The functional relevance of these SNPs remains to be determined. In monocytes of type 2 diabetic patients, the activity of membrane PKC and the membrane content of the PKCII isoform are acutely regulated by plasma glucose (284). In patients with diabetic nephropathy, the mesangial PKCII expression correlated with serum creatinine, interstitial macrophages, and interstitial fibrosis (285). 6. Agents with effects on DAG/PKC in diabetic kidney disease. The available pharmacological approaches to target PKC include

1. The DAG-PKC system. PKC, a family of serine-threonine kinases, consists of at least 12 structurally related isoforms (262, 263). The classical or conventional PKCs (, I, II, and ) require Ca2 and DAG to become activated, the novel PKCs (, , , , , and ) require only DAG, and the atypical PKCs, namely , , and (the mouse homolog of human PKC) require neither Ca2 nor DAG (262, 264). PKCs participate in signal transduction and intracellular communication in response to specific hormonal, neuronal, and growth factor stimuli. DAG, the major cellular mediator of PKC activation, can be derived from the hydrolysis of phosphoinositol biphosphate to inositol triphosphate or can be formed de novo from glycolytic intermediates (264). PKC isoform activation involves translocation by isozymespecific anchoring proteins, named receptors for activated C-kinase (265). PKC activation leads to changes in vascular permeability, ECM synthesis, smooth muscle contraction, gene expression, cell growth, differentiation, and angiogenesis (264, 266). 2. Expression of DAG/PKC in the normal kidney. The expression of PKC isoforms varies markedly between cells and tissues. The PKC, -I, -II, -, -, -, - isoforms were expressed in normal kidney; PKC was absent; and the expression of the -, -, /-isoforms was unknown (262, 267). However, another study demonstrated PKC, -, -, -, -, -, -, -, -, and - isoforms to be present in preglomerular vessels and glomeruli of normal rat kidney (81, 268). Furthermore, PKC, -I, -II, -, -, and - isoforms were identified by immunoblotting in vascular endothelial cells (269). PKC, -, -, and - were all expressed in rat kidney: PKC in cortical and outer medullary collecting ducts; PKC in thick ascending limbs and inner medullary collecting ducts; PKC and PKC were present in all nephron segments but with a different level of expression (270). In adult kidneys, mesangial cells express PKC and -I, whereas PKCII staining was found only in parietal epithelial cells (81, 271). 3. In vitro evidence for DAG/PKC effects on renal cells. PKC is activated in cultured mesangial cells (272, 273) and in explants of rat glomeruli (274, 275) exposed to elevated glucose concentrations. Hyperglycemia preferentially led to the activation of the PKC and -I isoforms in glomerular cells

Downloaded from edrv.endojournals.org on April 19, 2006

986

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

PKC inhibitors, AS-ODNs, and ribozyme inhibition of PKC (263). Staurosporine and several indolocarbazole derivatives are PKC inhibitors with a poor specificity (263). Therefore, in vivo studies have not been feasible until more specific PKC inhibitors were developed. Ruboxistaurin (LY333531) mesylate, a bisindolylmaleimide and highly specific PKCI/II inhibitor, is one of the most promising isoform-selective inhibitors developed to date. Other strategies like vitamin E and thiazolidinediones (TZDs) have also been shown to block PKC activity. a. AS-ODNs. In vitro, AS-ODNs against PKC reduced the high glucose-induced endothelial cell permeability, whereas the control sense and scrambled ODN and the ODN against PKC and - had no effect (286). In vivo, mice injected ip with ODN demonstrated a dose-dependent reduction in PKC mRNA. Studies evaluating the use of AS-ODNs against PKC isoforms for the treatment of diabetes-induced changes in the rat are under way (286).

b. PKC inhibitors. LY333531 decreased the high glucoseinduced PKC activity without changing the increased DAG levels in rat mesangial cells (272). Moreover, in human mesangial cells, LY333531 attenuated PKC activation and normalized the high glucose-induced changes in arachidonic acid release, PGE2 production, and NaK ATPase activity (272). In STZ-diabetic rats, treatment with LY333531 for 2 wk normalized elevated PKC activity in diabetic rat glomeruli, reduced elevated UAE, and normalized the elevated GFR of diabetic rats (281). Treatment with LY333531 for a longer period (12 wk) also normalized the overexpression of TGF1, fibronectin, and collagen 1 (IV) (272). Recently, treatment with LY333531 for 16 wk in db/db mice normalized the increased glomerular PKC activity, reduced the increase in UAE (Fig. 7A) and mesangial area (Fig. 7B), and reduced the overexpression of TGF-1, fibronectin, and type IV collagen (282). In the STZ-diabetic (mRen-2)27 rat, treatment with LY333531 for 6 months attenuated albuminuria, glomerulosclerosis, and, to a lesser extent, tubulointerstitial fibrosis

FIG. 7. A, Effect of PKC inhibition on the UAE rate of nondiabetic db/m and diabetic db/db mice. A 24-h urine sample for each mouse was collected in metabolic cages on 2 consecutive days 16 wk after the start of this experiment. Urine samples were processed to measure urinary albumin concentration using a competitive ELISA. Data were the average of UAE of 2 consecutive days and were shown as mean SD from six to nine mice. Numbers of mice from each group were denoted below each column. *, P 0.05 vs. other group. Quantification of glomerular pathology. Sections stained with periodic acid-Schiff were coded and read by an observer unaware of the experimental protocol. In each animal of four experimental groups, 20 glomeruli cut at the vascular pole were used for morphometric analysis. The extent of increase in mesangial matrix was determined by the presence of periodic acid-Schiff -positive and nuclei-free area in the mesangium. The glomerular area was also traced along the outline of capillary loop using a computer-assisted color image analyzer. Relative glomerular area was shown as the ratio of mesangial area/glomerular area. B, Mesangial area; C, glomerular area; D, relative mesangial area. Numbers of mice from each group are denoted below each column. *, P 0.01 vs. other group. **, P 0.05 vs. db/m and db/m treated with PKC inhibitor. [Reproduced with permission from D. Koya et al.: FASEB J 14:439 447, 2000 (282). The Federation of American Societies for Experimental Biology.]

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 987

(287). In a study in STZ-diabetic rats, LY333531 showed renoprotective effects without effect on any components of the intrarenal TGF- system (288). A clinical trial to evaluate whether LY333531 will be an effective treatment for diabetic nephropathy in combination with ACE inhibition or ARB therapy is ongoing (289). c. Vitamin E. In vitro, vitamin E suppressed DAG accumulation in thrombin-stimulated endothelial cells (290) and prevented the hyperglycemia-induced activation of PKC in VSMC (291) through the activation of DAG kinase (290, 291), which lowers intracellular DAG levels by converting DAG to phosphatidic acid. In STZ-diabetic rats, vitamin E prevented the diabetes-induced increases in DAG and PKC activity in rat glomeruli, normalized glomerular hyperfiltration, and improved UAE, partly through enhancing DAG kinase activity (292). It remains to be determined whether vitamin E directly affects DAG kinase activity or affects it indirectly through its antioxidant effect. d. TZDs. Although the prevention of renal complications by troglitazone (7) and pioglitazone (293) in type 2 diabetic rats is most likely due to their long-term positive effects on the glucose and lipid metabolism, other evidence suggests that TZDs may also have a direct effect on glomerular pathophysiology. In cultured mesangial cells, troglitazone and pioglitazone prevented the high glucose-induced activation of the DAG-PKC pathway activating DAG kinase (273), and troglitazone suppressed the secretion of type I collagen (294). Troglitazone also enhanced DAG kinase activity in glomeruli of diabetic rats and prevented glomerular hyperfiltration and albuminuria without changing blood glucose levels. TZDs are potential therapeutic agents for diabetic nephropathy that may prevent glomerular dysfunction independent of their insulin-sensitizing action through the inhibition of the DAG-PKC-ERK pathway (273). e. Others. Epalrestat abolished the glucose-induced enhancement of PKC activity in cultured mesangial cells (82); aminoguanidine and ramipril prevented the diabetes-associated increase in glomerular PKC activity (49), suggesting a link between the polyol pathway, AGE accumulation, RAS, and the PKC pathway, respectively. 7. Conclusion. In cell cultures and experimental type 1 and type 2 diabetes, hyperglycemia stimulates PKC activity, mostly PKC and PKCI, in a direct way or through an increase in DAG. This PKC activation stimulates prostaglandin, cytokine, and ECM protein production and various signal transduction molecules and decreases NaK-ATPase activity. Preliminary clinical evidence indicates a role for mesangial PKCII in diabetic nephropathy. Selective PKCII inhibition normalized glomerular hyperfiltration, reduced UAE, and decreased glomerular TGF-1 expression and ECM accumulation in experimental diabetes, hence reducing mesangial expansion, glomerulosclerosis, tubulointerstitial fibrosis, and loss of renal function. Although these findings clearly implicate a role for PKC in these diabetes-induced changes, the role of other PKC isoforms cannot be excluded.

V. Growth Factors and Cytokines A. Transforming growth factor (TGF-)

1. The TGF- system. The TGF- system belongs to the TGF- superfamily of multifunctional cytokines (295). The three mammalian isoforms, TGF-1, TGF-2, and TGF-3, strongly induce ECM synthesis. TGF-s also contribute to embryonic development, tissue homeostasis, hematopoiesis, angiogenesis, chemotaxis, and immune functions. TGF-s are synthesized and secreted as a latent high-molecular weight complex consisting of mature TGF-, latency-associated peptide, and a latent TGF- binding protein. After proteolytic activation, biologically active TGF-1, TGF-2, and TGF-3 homodimers are most abundantly generated (295). Three TGF- receptors have been identified, i.e., two serine/threonine kinase receptors, TGF- type I receptor (TGF- RI) and TGF- type II receptor (TGF- RII) (296); and TGF- RIII, which facilitates the binding of TGF-s to TGF- RII (295). TGF- signal transduction involves three subclasses of Smad proteins, i.e., receptor-activated (R-Smads), common-partner (Co-Smads) and inhibitory Smads (ISmads), and another pathway, the TGF--activated kinase 1/MAPK signaling pathway (295, 297). Recently, CTGF, a novel fibrogenic protein, has been identified as another downstream mediator of TGF- (298). 2. Expression of TGF- in the normal kidney. The kidney is a site of TGF- production and a target of TGF- action, because both mRNA for all three TGF- isoforms and receptors and the active TGF- proteins have been identified in all cell types of the glomerulus and in proximal tubular cells (299, 300). TGF-1, TGF-2, and TGF-3 differ, however, in their in vivo expression patterns (295). In situ hybridization of rat renal tissue revealed sparse hybridization for TGF-1 mRNA in the tubulointerstitium and a stronger TGF-1 gene expression within proximal tubular cells (301). In mice, the TGF-1 protein was localized in the proximal and distal tubules rather than the glomeruli (302). In rat kidney, podocytes expressed TGF- RIII and very occasionally TGF- RII, but not TGF- RI, whereas all three receptors were constitutively expressed on glomerular endothelial cells (303). CTGF was expressed in cultured mesangial and proximal tubular cells (298, 304). CTGF expression was reported in cortical distal tubules and in medullary and papillary collecting ducts in rat kidney (304), and in glomeruli of mice (298). 3. In vitro evidence for TGF- effects on renal cells. High glucose (305) and glycated albumin (27) stimulated TGF-1 production in cultured mesangial and glomerular endothelial cells. In addition, in cultured mesangial cells, high glucose- or Ang-stimulated matrix protein production was partly mediated by TGF- (306, 307), and TGF- modulated the effect of high glucose on NO production (225). Potential mechanisms for the increased ECM production by TGF-1 in glomerular mesangial, glomerular epithelial, and tubular cells include inhibition of MMP synthesis, stimulation of metalloproteinase inhibitor production, increased expression of PDGF-BioBreeding (BB), CTGF, monocyte chemoattractant protein-1, and regulated upon activation, normal T cell expressed and secreted (RANTES) (298, 308 312). In vitro,

Downloaded from edrv.endojournals.org on April 19, 2006

988

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

CTGF induced migration of mesangial cells and increased fibronectin and collagen type I secretion (298, 313). Furthermore, high glucose-induced CTGF mRNA expression and protein secretion in mesangial cells was mediated by TGF-, as shown by its inhibition by an anti-TGF- neutralizing antibody (298). 4. Experimental evidence for a role of TGF- in diabetic kidney disease. In experimental type 1 diabetes, glomerular and tubular TGF-1 mRNA was increased early in the course of diabetes-induced renal changes (301, 314 317). In addition, the increase in glomerular TGF-1 mRNA was sustained in long-term STZ-diabetic rats (300). Recently, the whole intrarenal TGF- system (i.e., TGF-1, TGF-2, and TGF-3 isoforms and TGF- type RI, RII, and RIII receptors) has been studied rigorously in acute and chronic type 1 diabetes, more particular, in STZ-diabetic rats and BB rats, a model of autoimmune type 1 diabetes (299). The TGF-1 and TGF-2 isoforms and the TGF- type RII were the most responsive elements to diabetes induction (299). In the acute phase of STZ diabetes, TGF-1 protein was decreased in the glomerulus but increased in tubules. Glomerular TGF-2 and TGF- type RII protein were up-regulated along with procollagen-1 C-propeptide, a marker for the rate of fibrosis (299). Recently, in db/db mice, in situ hybridization and immunohistochemical staining revealed increases in glomerular and tubular TGF-1 and TGF- RII mRNA and protein when compared with their nondiabetic littermates (302). In OLETF rats, TGF-1 immunostaining was increased compared with Long-Evans Tokushima Otsuka control rats, particularly in interstitial cells and ECM in fibrotic interstitial lesions, and in atrophic or dilated tubular cells, whereas normal tubular cells and glomeruli only stained weakly (318). Another study observed increased kidney TGF-1 mRNA in OLETF rats compared with controls, but TGF-1 immunostaining was different from the previous study, with positive staining especially in the glomerular mesangial area, and weak staining in the tubules (319). Interestingly, administration of recombinant human TGF-2 in normal rats did not induce renal hemodynamic changes, and very little fibrosis was observed (320). CTGF mRNA levels were increased in the renal cortex of STZ-diabetic rats compared with controls, particularly in dilated-appearing proximal tubules, in which it tended to colocalize with IGF-I (304). In the early phase of diabetic kidney disease in db/db mice, when mesangial expansion was mild and interstitial disease and proteinuria were absent, glomerular CTGF expression was increased 27-fold when compared with control mice (298). In whole kidney cortices, a substantially lower elevation of CTGF mRNA was observed, indicating that the primary alteration of CTGF expression was in the glomerulus (298). 5. Clinical evidence for a role of TGF- in diabetic kidney disease. Normoalbuminuric type 1 diabetic patients with a diabetes duration of more than 2 yr had lower serum TGF- levels than control subjects (321). Only 38% of young type 1 diabetics with normo- or microalbuminuria had higher urinary TGF-1 excretion than control subjects (322). The urinary TGF-1 correlated weakly with urinary concentrations of glucose and 1-microglobulin, a marker for tubular dysfunc-

tion, but not with UAE (322). Furthermore, glycemic control did not predict urinary TGF-1 levels in type 1 diabetic subjects (323). The association of urinary TGF-1 excretion with diabetic nephropathy remains controversial (322, 323). Increased TGF- immunostaining has been described in glomeruli and tubulointerstitium of type 1 diabetic subjects with nephropathy (324, 325), but has also been reported in other renal diseases characterized by ECM accumulation (324). Furthermore, a positive correlation between TGF-, fibronectin, and plasminogen activator inhibitor-1 levels in glomeruli and tubulointerstitium was found (324). In normoalbuminuric type 2 diabetic patients, serum TGF- levels were higher than in control subjects irrespective of the diabetes duration (321). In type 2 diabetic patients, an increased renal production of TGF-1 has been shown by measuring aortic, renal vein, and urinary levels of TGF-1 (326). The increased urinary TGF-1 excretion (326 328) correlated with UAE (327) and was associated with severe mesangial expansion (328). However, both renal impairment and diabetes may independently lead to increased urinary TGF-1 levels (329). A higher expression of TGF-1 mRNA was found in glomeruli of type 2 diabetic patients with diabetic nephropathy as compared with normal subjects, and intraglomerular TGF-1 mRNA levels correlated with hemoglobin A1c levels (330). Examination of kidney biopsy specimens for CTGF mRNA by in situ hybridization showed up-regulation of glomerular CTGF mRNA in lesions of diabetic nephropathy. However, this is not specific for diabetic nephropathy because it was also demonstrated in lesions of other renal diseases (331). 6. Agents with effects on the TGF- system in diabetic kidney disease. Several substances are able to inhibit the action of TGF-s, i.e., neutralizing antibodies, AS-ODNs, and dominant negative mutant TGF- receptors. Specific targeting of CTGF is possible with CTGF antibodies and AS-ODNs, but to date no studies have been published regarding the influence of CTGF inhibition in diabetic nephropathy. Other classes of drugs have also been shown to affect the expression of the TGF- system in diabetes, including ACEi, PKC inhibitors, statins, and inhibitors of AGE formation. a. Neutralizing antibodies. In vivo, short-term (9 d) ip administration of a pan-neutralizing TGF- antibody in STZdiabetic mice attenuated the increased renal TGF-1 and TGF- RII mRNA levels and reduced both the diabetesassociated renal/glomerular growth and enhanced renal expression of type IV collagen and fibronectin (332). Systemic treatment for 14 d with recombinant human monoclonal anti-TGF-2 IgG4 (termed CAT-152) in STZ-diabetic rats attenuated the rate of type I collagen synthesis and reduced UAE levels compared with placebo-treated diabetic rats (333). In addition, chronic inhibition of the biological actions of TGF- with a pan-neutralizing monoclonal antibody in db/db mice decreased total plasma TGF-1 levels, attenuated the increase in plasma creatinine concentrations, and substantially attenuated the increase in renal type IV collagen and fibronectin mRNAs, thus preventing glomerulosclerosis (Fig. 8) (334). Interestingly, this study did not find any effect of TGF- antibodies on UAE (334).

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 989

FIG. 8. Kidney matrix gene expression and mesangial matrix expansion in diabetic mice treated with anti-TGF- antibody. Summary of densitometric analyses of 1(IV) collagen/mrpL32 mRNA ratios (A) and fibronectin/ mrpL32 mRNA ratios (B) in the four treatment groups (mean SE; n 8 for each IgG group, and n 9 for each T group). The relative mRNA ratio in the normal-IgG group is assigned a value of 1. *, P 0.05 vs. normal IgG; **, P 0.05 vs. diabetic IgG. Quantitative measurement of extracellular mesangial matrix expansion (C) is expressed as periodic acid-Schiff-positive mesangial material per total glomerular tuft cross-sectional area. An average value was obtained from analyses of 30 glomeruli per mouse. Data are mean SEM; n 8 for each IgG group, and n 9 for each T group. *, P 0.05 vs. normal IgG; **, P 0.05 vs. diabetic IgG. [Reproduced with permission from F. N. Ziyadeh et al.: Proc Natl Acad Sci USA 97:8015 8020, 2000 (334). National Academy of Sciences of the United States of America.]

b. TGF-1 AS-ODNs. In vivo, treatment of STZ-diabetic mice for 10 d with AS-ODNs via osmotic minipumps partially decreased kidney TGF-1 protein levels and prevented the increase in kidney weight and ECM accumulation that were seen in placebo-treated diabetic mice (335). c. ACEi. The effect of ACE inhibition on the intrarenal changes of all three TGF- isoforms and receptors has been examined in experimental type 1 diabetes (336). Enalapril reduced the diabetes-associated renal hypertrophy, albuminuria, and up-regulation of TGF- receptors but had no effect on the glomerular expression of the TGF- isoforms (Fig. 9) (336). Furthermore, in STZ-diabetic rats, administration of ramipril prevented the tubulointerstitial injury and the renal tubular overexpression of TGF-1 and 1 (IV) collagen mRNA (301). In type 1 diabetic patients, captopril treatment for 6 months lowered serum TGF-1 levels. The fall in serum TGF-1 correlated with the ACEi-induced stabilization of GFR over a 2-yr period in patients with overt nephropathy (337). d. PKC inhibition. The specific PKC isoform inhibitor LY333531 abolished the diabetes-associated increases in glo-

merular TGF- mRNA and ECM accumulation when given to STZ-diabetic rats for 3 months (272). In a long-term study in db/db mice, PKC inhibition resulted in the reduction of long-term renal changes, including the increase in glomerular TGF-1 immunostaining (282). e. Statins. Lovastatin suppressed the high glucose-induced up-regulation of TGF-1 and fibronectin mRNA and proteins in cultured rat mesangial cells (338) and suppressed the increase in UAE, kidney weight, glomerular volume, and glomerular TGF-1 mRNA expression in STZ-diabetic rats (338). Simvastatin inhibited CTGF mRNA expression in a concentration-dependent way in cultured human mesangial cells (339). f. AGE inhibition. Long-term treatment with the inhibitor of AGE formation OPB-9195 in OLETF rats normalized the diabetes-associated increases in renal TGF-1 expression and type IV collagen accumulation and decreased albuminuria (319). g. ET receptor antagonists. The mRNA levels of TGF-, PDGF-B, TNF-, basic fibroblast growth factor (bFGF), and

Downloaded from edrv.endojournals.org on April 19, 2006

990

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

FIG. 9. Data for procollagen-I C-propeptide (A), TGF-1 mRNA (B), and TGF- type II receptor (TGF-RII) mRNA (C) at d 30 in nondiabetic controls (black bars), placebo-treated diabetic rats (open bars), and enalapriltreated diabetic rats (gray bars). All densitometry in arbitrary units SEM, n 3. *, P 0.05, placebo-treated diabetic rats vs. nondiabetic control rats; **, P 0.05, enalapril-treated diabetic rats vs. placebo-treated diabetic rats. [Reproduced with permission from C. Hill et al.: Diabetologia 44:495500, 2001 (336). Springer.]

of collagen I, III, and IV, laminin B1 and B2 all increased with age in glomeruli of STZ-diabetic rats (192). Twenty-four weeks of treatment with the ETA antagonist FR139317 attenuated the increases in the glomerular mRNA levels of these growth factors and ECM components, attenuated the rise in CrCl, and reduced urinary protein excretion in diabetic rats without any effect of FR139317 in glomeruli of control rats. h. Others. The antioxidant d--tocopherol blocked the high glucose-induced increases in TGF- and matrix accumulation in cultured mesangial cells (340) and prevented the increase in glomerular TGF- immunoreactivity and glomerular size in STZ-diabetic rats (341). The TZD troglitazone prevented not only diabetic glomerular hyperfiltration and albuminuria, but also the increase in mRNA expression of ECM proteins and TGF-1 in glomeruli of diabetic rats (273). Administration of the oral adsorbent AST-120 to OLETF rats attenuated the progression of glomerular sclerosis, tubulointerstitial fibrosis, as well as renal dysfunction. AST-120 reduced the interstitial expression of TGF-1 and tissue inhibitor of metalloproteinase-1 (318). Furthermore, combination therapy of AST-120 and benazepril was more effective than benazepril alone in retarding the progression of interstitial fibrosis by reducing the expression of TGF-1, tissue inhibitor of metalloproteinase-1, and osteopontin (342). 7. Conclusion. In vitro evidence clearly demonstrates that glucose up-regulates TGF- and CTGF, which can induce the expression of ECM proteins. Furthermore, TGF- can also induce ECM production indirectly by stimulating PDGF and chemokines. Compelling evidence demonstrates up-regulation of renal TGF-1, TGF- RII, and CTGF and, to a lesser extent, of TGF-2, in experimental type 1 and type 2 diabetes. Similarly, up-regulation of renal TGF-1 and CTGF and increased urinary TGF-1 excretion have been documented in type 1 and type 2 diabetic patients. The extended experimental and clinical studies indicate that TGF- and CTGF, as

fibrogenic factors, with CTGF acting downstream of TGF-, are important factors in the pathogenesis of diabetic nephropathy, i.e., in mesangial matrix accumulation and in tubulointerstitial fibrosis. Urinary excretion of TGF-1 seemed to correlate with UAE or tubular dysfunction. Inhibition of TGF- by neutralizing antibodies or AS-ODNs attenuated the up-regulation of TGF-1 and reduced renal and glomerular growth and the expression of ECM proteins in experimental type 1 and type 2 diabetes, giving further evidence for a strong pathogenetic role of TGF- in the structural alterations of diabetic kidney disease. The antibodies also reduced UAE in type 1 diabetic and plasma creatinine levels in type 2 diabetic animals, suggesting that TGF- may also play a minor role in diabetic renal dysfunction. Unfortunately, no agents that directly affect the TGF- system exist for clinical use in diabetic patients, and therefore the experimental results cannot be validated in human studies.
B. Growth hormone (GH) and insulin-like growth factors (IGFs)

1. The GH/IGF system. The GH/IGF system consists of a complex family of peptides in the circulation, extracellular space, and in most tissues. GH classically induces IGF-I synthesis in various organs through activation of specific GH receptors (GHRs). Recently, three isoforms of the GHR, i.e., the full-length GHR, GHR-(1279), and GHR-(1277), have been identified in human tissues (343). The GHR signals through receptor-associated Janus-activated kinase 2, signal transducer and activator of transcription proteins, Ras/ MAPK, and phosphatidylinositol-3-kinase. The GHR is down-regulated by internalization and degradation and by phosphatases or suppressors of cytokine signaling proteins (344). The GH binding protein (GHBP) may serve as a circulating buffer/reservoir function for GH, prolong the plasma GH half-life, compete with GH for GHRs, and form unproductive heterodimers with the GHR. The net effect of

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 991

these partly enhancing and partly inhibitory functions on GH action in vivo is complex and difficult to ascertain (345). The IGF system consists of IGF-I and IGF-II, IGF-I receptor (IGFIR), IGF-II/mannose-6-phosphate receptor (IGF-II/man-6PR), IGF binding proteins (IGFBPs), and IGFBP proteases (346). The mitogenic effects of IGF-I on cell growth and metabolism are mediated mainly through the IGF-IR, which binds IGF-I and IGF-II with high affinity (347). The IGF-IIR/ man-6-PR binds IGF-II and mannose-6-phosphate-containing ligands and plays a role in trafficking of lysosomal enzymes (348, 349). In the kidney, GH and IGF-I play a role in renal hemodynamics and in tubular phosphate, sodium, and water absorption (350). IGFs are normally bound to highaffinity IGF binders, IGFBP-1 to IGFBP-6, and several lowaffinity IGF binders, IGFBP-related proteins (IGFBP-rP1 to IGFBP-rP9), and IGFBP proteolytic fragments (346). IGFBPrP2 is identical to CTGF (346) and has been discussed in Section V.A. Circulating IGFBPs may regulate the half-life and endocrine effects of IGFs, whereas cellular IGFBPs may inhibit or stimulate local actions of IGF. The physiological role of the IGFBP-rPs remains to be determined. 2. Expression of GH/IGFs in the normal kidney. The GH/IGF system is expressed in the normal kidney, i.e., GHR and GHBP, IGF-I and IGF-II, the respective IGF-IR and IGF-II/ man-6-PR, and all six specific IGFBPs (343, 345, 346, 348 350). All three GHR isoforms have been discovered in human kidney (343). In rat kidney, GHR mRNA was detected most abundantly in the proximal tubule, less in the medullary thick ascending limb, and not at all in the glomerulus (351), whereas IGF-IR mRNA was concentrated in the medullary thick ascending limb, the distal nephron and collecting duct, and in the glomerulus, with the lowest levels in the proximal tubules (352, 353). IGF-I staining was confined to a few collecting ducts (352). The renal glomerulus is a site of both action and synthesis of IGF-I. Mouse glomerular mesangial cells synthesize and release IGF-I (354), and specific IGF-I receptors are present in cultured glomerular endothelial, epithelial, and mesangial cells (355357), suggesting an autocrine and paracrine action of IGF-I in the kidney. 3. In vitro evidence for GH/IGF effects on renal cells. IGF-I is a potent mitogen for glomerular mesangial cells (355, 358). IGF-I stimulated proteoglycan production (359), as well as the production of laminin, fibronectin, and type IV collagen in mesangial cells (358). Recently, autocrine activation of the IGF-I signaling pathway has been demonstrated in mesangial cells isolated from NOD mice (305). Mesangial cells isolated from obese type 2 diabetic db/db mice exhibited higher levels of IGF-I receptors compared with cells from nondiabetic littermates (360). IGF-I induces NO synthesis and release by cultured vascular endothelial cells through IGF-I receptors (350). 4. Experimental evidence for a role of GH/IGFs in diabetic kidney disease. Experimental evidence for a role of GH/IGFs in the development of diabetic kidney disease is quite extensive (361). Nondiabetic transgenic mice expressing GH, GH releasing factor, or IGF-I, exhibit important glomerular enlargement (362). The GH or GH releasing factor transgenic mice developed glomerulosclerosis, indicating that the mes-

angial changes were in part due to circulating GH (362). Furthermore, glomerular size in GH transgenic mice correlated with UAE and the degree of glomerulosclerosis (363). The STZ-induced diabetic rat, a model of type 1 diabetes, is characterized by low circulating GH concentrations (364), in contrast to findings in humans (365) and STZ-diabetic mice (361). The reason for this discrepancy is still controversial. However, the difference seems to be restricted to GH because similar changes for other elements in the GH/IGF axis have been reported, including low circulating levels of GHBP and IGF-I, alterations in serum IGFBP levels, and specific changes in renal GHBP, IGF receptors, and IGFBPs (361, 364, 366 370). Renal GHBP mRNA expression was increased both in short-term and long-term diabetic rats, but not accompanied by a change in GHR mRNA expression (371). The initial increase in renal growth and function in experimental diabetes was preceded by a rise in renal IGF-I (369, 372), IGFBPs (348, 373), and IGF-II/man-6-PR concentration (374). Furthermore, specific changes occurred in the renal GHBP mRNA, IGF-IR mRNA, and IGFBP mRNA expression in long-term experimental diabetes (368). IGF-I immunostaining increased early in STZ-diabetic rats, in cortical collecting ducts, the thick ascending limbs of the loops of Henle, the macula densa, and some distal convoluted tubules, suggesting that IGF-I is involved in diabetic tubular injury (352). Substantial experimental evidence suggests that exogenous and endogenous IGF-I raises RPF and GFR by reducing renal arteriolar resistance and increasing the glomerular ultrafiltration coefficient, possibly by relaxing the mesangium. These effects of IGF-I on renal hemodynamics are mediated through IGF-I receptors and by induction and release of NO. GH probably does not directly influence renal hemodynamics but increases GFR and RPF through the induction of IGF-I (350). Prepubertal diabetic rats have reduced kidney growth and kidney IGF-I levels compared with adult diabetic rats (375). STZ-diabetic dwarf rats with isolated GH and IGF-I deficiency showed less renal and glomerular hypertrophy and a smaller rise in UAE than diabetic control rats with intact pituitary (376, 377). 5. Clinical evidence for a role of GH/IGFs in diabetic kidney disease. Recently, various clinical studies have been published dealing with possible correlations between serum or urinary GH/IGF-I levels and kidney function in type 1 diabetic patients. Prepubertal microalbuminuric patients had higher levels of urinary GH and urinary and plasma IGF-I than normoalbuminuric diabetic and control subjects (378). Prepubertal and pubertal circulating IGF-I levels were positively correlated to GFR, but not to UAE (378, 379). In adult normoand microalbuminuric patients, urinary IGF-I was strongly correlated to kidney volume, and both urinary IGF-I and GH were positively correlated to microalbuminuria (380). In contrast, no correlations between either plasma GH or IGF-I concentrations and GFR were found in normoalbuminuric type 1 diabetic patients (381). Another study found higher serum GH levels and lower plasma IGF-I levels in type 1 diabetic patients compared with controls, but no correlation between plasma IGF-I and renal function (382). A crosssectional study reported lower IGF-I and IGFBP-3 but higher IGFBP-1 levels in type 1 diabetics compared with type 2

Downloaded from edrv.endojournals.org on April 19, 2006

992

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

diabetic patients or controls (383). IGFBP-5 levels were lower in both diabetic groups than in controls, whereas IGFBP-4 levels were similar in diabetics and controls (383). Finally, urinary IGFBP-3 proteolysis correlated with UAE in type 1 and type 2 diabetic subjects (384). Whether induction of IGFBP-3 proteolysis contributes to diabetes-induced renal structural changes by increasing IGF bioavailability is currently unknown (384). 6. Agents with effects on the GH/IGF system in diabetic kidney disease. Various agents have been shown to affect the GH/ IGF axis and exert beneficial renal effects in diabetic kidney disease, including substances that directly target the GH/ IGF axis, such as long-acting somatostatin analogs, specific GHR antagonists, and IGF-I receptor antagonists, and substances that indirectly influence the GH/IGF axis by targeting other systems such as inhibitors of AGE formation and ACEi. a. Long-acting somatostatin analogs. Somatostatin analogs suppress elevated circulating GH levels. In short-term experimental diabetes in the rat, treatment with octreotide from diabetes onset completely inhibited the initial renal hypertrophy and kidney IGF-I accumulation (385), whereas postponed treatment resulted in a partial inhibition of renal hypertrophy (386). Six months of octreotide treatment from the induction of diabetes reduced the increases in kidney weight, renal IGF-I levels, and UAE when compared with untreated rats (387). Octreotide treatment for 3 wk following 3 months of untreated diabetes reduced kidney weight but had no effect on kidney IGF-I levels or UAE. However, the combined treatment of octreotide and captopril reduced both kidney weight and UAE (4). In a type 1 diabetic mouse model, octreotide prevented renal and glomerular growth partly through inhibition of GH hypersecretion and of local kidney IGF-I levels (388). Recently, a new somatostatin analog, PTR3173, blunted renal/glomerular hypertrophy, albuminuria, GFR and renal IGF-I accumulation in NOD mice (389). In clinical studies, acute infusion of octreotide to type 1 diabetic patients reduced RPF, GFR, plasma GH, and glucagon levels (390). In addition, continuous sc octreotide infusion for 12 wk in type 1 diabetic patients reduced elevated GFR and kidney volume (391). b. GHR antagonists. Diabetic GHR antagonist transgenic mice and diabetic GHR/GHBP knockout mice are protected against the development of diabetic renal changes (392394). Furthermore, sc injections of a GHR antagonist, G120K-PEG, for 4 wk normalized the diabetes-associated increases in renal IGF-I accumulation, kidney weight, and glomerular volume and attenuated the diabetes-associated rise in UAE in STZ-diabetic mice and NOD mice without affecting circulating GH and IGF-I levels (Fig. 10) (369, 395). GHR antagonist treatment was equally potent to ACEi treatment in reducing UAE (369). Clinical studies on the effects of this new group of GHR antagonists in diabetic patients may be initiated in the years to come. c. IGF-I receptor antagonists. Treatment with JB3, a peptide analog of IGF-I, for 4 d in STZ-diabetic rats normalized the diabetes-induced increase in kidney weight, kidney protein, and DNA (396).

FIG. 10. Mean glomerular volume (left) and UAE (right) at d 28 in nondiabetic mice treated with placebo (CP) or G120K-PEG (CA) and diabetic mice treated with placebo (DP) or G120K-PEG (DA). Values are means SEM; n 10 in each group. *, P 0.05 vs. all other groups (CP, CA, and DA). , P 0.05 vs. all other groups (CP, CA, and DP). [Reprinted with permission from A. Flyvbjerg et al.: Diabetes 48:377 382, 1999 (369). American Diabetes Association.]

d. ACEi. The possible effects of ACEi on the GH/IGF axis in diabetic kidney disease have not been examined extensively. In a short-term experimental study in STZ-diabetic rats, trandolapril treatment for 1 wk had no effect on kidney IGF-I accumulation or renal enlargement (397). As mentioned above, monotherapy with captopril or octreotide had no effect on renal IGF-I concentrations or UAE, although kidney weight and UAE were reduced with combination therapy (4). e. AGE inhibition. A recent study in long-term STZ-diabetic rats showed the classic changes in the intrarenal IGF axis, with decreased IGF-I and IGFBP-4 and increased IGFBP-1 mRNA expression (398). Administration of aminoguanidine (see also Section II. A) normalized renal IGFBP-1 mRNA expression and partially restored the diabetes-associated changes in IGFBP-4 and IGF-I mRNA (398). 7. Conclusion. Ample in vitro and experimental evidence indicates a role for the GH/IGF-I axis through the complexity of GHR, GHBP, IGFs, IGF receptors, and IGFBPs in both early and late renal changes in experimental diabetes. IGF-I, in particular, seems responsible for the early renal structural and functional changes in diabetic kidney disease, i.e., in renal and glomerular hypertrophy, mesangial hyperplasia, increased UAE and GFR, and tubular injury and growth. Specific changes in the GH/IGF-I system in long-term experimental diabetes seem to involve primarily IGFBPs and receptors. In diabetic patients, in general, circulating GH levels are increased and IGF levels low. A full characterization, however, of the different components of the GH/IGF axis in human diabetic kidney disease awaits future studies. Somatostatin analogs lower circulating GH levels, whereas GHR antagonists prevent functional GHR dimerization and lower circulating concentrations of IGF-I. Despite their different way of action, both treatments had clear beneficial effects on renal and glomerular growth, renal IGF-I accumulation, and UAE in experimental diabetes. In diabetic patients, however, somatostatin analogs mainly reduced the increase in GFR and kidney volume but had no effect on

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 993

UAE. Therefore, clinical use of these agents in the treatment of diabetic kidney disease will depend largely on the outcome of future studies.
C. Vascular endothelial growth factor (VEGF)

1. The VEGF system. The family of VEGFs currently includes VEGF-A, -B, -C, -D, -E, and placenta growth factor (399). VEGF-A, or VEGF, exists as at least six different homodimeric glycoproteins of 121, 145, 165, 183, 189, and 206 amino acids (VEGF121206) in humans and one amino acid shorter in rodents (399, 400). VEGF stimulates endothelial cell proliferation and differentiation, increases vascular permeability, mediates endothelium-dependent vasodilatation, plays a cardinal role in physiological and pathological angiogenesis, and modulates leukocyte kinetics (400, 401). The two best-described VEGF receptors (VEGFR-1 and VEGFR2), also known as fms-like tyrosine kinase 1 and fetal liver kinase 1/KDR, are high-affinity transmembrane tyrosine kinase receptors (400). Soluble fms-like tyrosine kinase, a splice variant of VEGFR-1, regulates VEGF availability by binding VEGF in the circulation (402). Neuropilins serve as isoformspecific coreceptors for VEGF. VEGF production is regulated by several growth factors and cytokines such as TGF-, PDGF, IGF-I (399, 400). VEGF up-regulates the expression of NOS3 in endothelial cells and increases the production of NO (403). 2. Expression of VEGF in the normal kidney. Both VEGF and the two VEGFRs are expressed in the glomeruli and tubules of normal kidney (402, 404 407). Cultured rat and human mesangial cells express both mRNA of VEGF121, VEGF165, and VEGF189 and VEGF protein (407, 408). In rodent and human kidney, VEGF mRNA and/or protein was detected in glomerular podocytes, distal tubules, and collecting ducts, and to a lesser extent in some proximal tubules (402, 404, 406 409). In human glomeruli, variable patterns of VEGF expression were identified, but mostly, all three common isoforms, VEGF121, VEGF165, and VEGF189 were expressed (402). VEGFR-1 and VEGFR-2 were expressed in cultured rat and human mesangial cells (410 412) and cultured rat tubular epithelial cells (413) but not in cultured human podocytes (414). In rat kidney, VEGFR-2 was primarily expressed in glomerular endothelial cells, but also in distal convoluted tubules, collecting ducts and interstitial cells, whereas VEGFR-1 was detected as more diffuse in proximal and distal tubules (404, 413). In human kidney, VEGFR-1 and VEGFR-2 were predominantly localized to preglomerular, glomerular, and postglomerular endothelial cells (405, 406, 411). The expression of VEGFR-1, VEGFR-2, soluble VEGFR-1, and neuropilin-1 in isolated human glomeruli was also heterogenous (402, 414). Neuropilin-1 was detected in whole kidney, single glomeruli, cultured human glomerular podocytes (414), and mesangial cells (410, 411). 3. In vitro evidence for VEGF effects on renal cells. Cultured mesangial cells, glomerular endothelial cells, proximal and distal tubular cells are capable of producing VEGF (415 417). High glucose-induced VEGF production in cultured rat mesangial cells seems to be PKC-dependent (407, 418). AGEs, Ang II, and TGF-1 induced VEGF secretion in cultured

human mesangial cells (408, 416, 417, 419). TGF-1 downregulated VEGFR-2 in vascular endothelial cells (420). In endothelial cells in vitro, VEGF stimulated NF-B concentrations (421) and caused a dose-dependent increase of ACE, suggesting a synergistic relation between VEGF and the RAS (422). In cultured human mesangial cells, VEGF induced proliferation (411) and increased collagen synthesis and p42/ p44 MAPK activity (410). VEGF induced a proliferative and antiapoptotic response in cultured rat tubular epithelial cells (413). Furthermore, murine proximal tubular cells demonstrated augmented de novo protein synthesis and hypertrophy in response to VEGF (423). 4. Experimental evidence for a role of VEGF in diabetic kidney disease. Increased renal expression of VEGF and VEGFRs has been demonstrated in STZ-diabetic rats (404). The increase in VEGF mRNA and protein in glomerular podocytes, distal tubules, and collecting ducts observed after 3 wk of diabetes was sustained until 32 wk of diabetes. In contrast, the early increase in VEGFR-2 mRNA in glomerular and peritubular endothelial cells and interstitial cells, was transient and not observed after 32 wk (404). Early induction of VEGF at the onset of diabetes was also demonstrated in the renal tubular and vascular compartments of STZ-diabetic rats with superimposed hypertension (424) and in genetically diabetic BB rats, with higher amounts of VEGF protein and mRNA than in nondiabetic rats (425). Increased renal VEGF expression has also been described in experimental models of type 2 diabetes (319, 334, 426). In OLETF rats, renal VEGF mRNA and glomerular immunoreactivity were higher than in nondiabetic Long-Evans Tokushima Otsuka rats (319). However, VEGF mRNA expression increased only in the early period of diabetic nephropathy and not further with the duration of diabetes. Positive staining for VEGF was mainly found within the glomeruli (319). In ZDF rats, renal VEGF mRNA levels rose early in the course of diabetes until 7 months (426). At 9 months, when glomerulosclerosis was most pronounced, renal VEGF mRNA levels were reduced (426). In kidneys of db/db mice, VEGF mRNA was increased 2-fold when compared with nondiabetic littermates (334). 5. Clinical evidence for a role of VEGF in diabetic kidney disease. In type 1 diabetic patients, increased (427 429) and unaltered (430, 431) serum or plasma VEGF levels have been found compared with healthy controls. Various studies aimed to correlate circulating VEGF levels to parameters of metabolic control or the severity of diabetic nephropathy. Compared with patients without complications (428, 432) or with microalbuminuria (427), macroalbuminuric patients had higher circulating VEGF levels. However, in other studies, VEGF levels were similar between type 1 diabetics with and without micro- or macroalbuminuria (430, 431, 433). In some studies, correlations were observed between VEGF levels and glycemic control, the severity of diabetic nephropathy or the degree of UAE (427, 428). However, no such correlations were found in other studies (430, 431, 433, 434). Urinary VEGF excretion was not different between patients with diabetic nephropathy and controls (435). Two studies describing VEGF expression in renal biopsy specimens from patients with different kidney diseases included patients

Downloaded from edrv.endojournals.org on April 19, 2006

994

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

with diabetic nephropathy (436, 437). Glomerular VEGF expression was highest in the patients with mildest sclerotic changes (436, 437), notably strong in viable glomerular podocytes, and decreased or absent with increasing sclerosis (437). In type 2 diabetic patients, plasma VEGF levels were higher than in controls (438). Plasma VEGF concentration tended to rise with increasing UAE (438); however, no such correlation was reported in other studies (434, 439). In type 2 diabetic patients, plasma VEGF concentration was higher in patients with overt proteinuria than in patients with normo- or microalbuminuria but did not differ between normo- and microalbuminuric patients (407). Urinary VEGF excretion increased with the progression of diabetic nephropathy and correlated weakly with the levels of serum creatinine, CrCl, microalbuminuria, and proteinuria (407). VEGF was up-regulated in glomerular podocytes and distal tubular cells in biopsies with mild diabetic nephropathy, whereas in biopsies with advanced diabetic nephropathy, VEGF expression was decreased or absent in sclerotic glomeruli but extensive in tubules, especially the proximal segment (407). Tubulointerstitial VEGF and VEGF mRNA were lower in type 2 diabetic patients with severe diabetic nephropathy (440). 6. Agents with effects on the VEGF system in diabetic kidney disease. Several VEGF/VEGFR antagonists have been developed, including AS-ODNs, soluble VEGFRs, VEGF aptamers, and ribozymes. In addition, VEGF receptor tyrosine kinase inhibitors, VEGFR-2 antagonists, and peptides that block the interaction of VEGF with its receptors are available. Finally, monoclonal antibodies directed against VEGF or its receptors have been produced. So far, only anti-VEGF antibodies have been used to study the role of VEGF in diabetic kidney disease. a. Neutralizing antibodies. Two studies have investigated the effects of anti-VEGF antibodies in experimental diabetic kidney disease. Treatment for 6 wk in STZ-diabetic rats with a monoclonal neutralizing VEGF antibody abolished the di-

abetes-associated increases in GFR, glomerular hypertrophy, and UAE (Fig. 11) (261). In addition, the diabetes-associated up-regulation of NOS3 expression was prevented. No effect on metabolic control was seen in diabetic animals, and no renal effects were seen in nondiabetic controls (261). Furthermore, VEGF antibody administration in db/db mice attenuated the diabetes-associated increases in kidney weight, glomerular volume, and UAE and abolished the increase in BMT and CrCl. In addition, VEGF antibody tended to reduce the expansion of total mesangial volume (Fig. 12) (441). A role for VEGF in glomerular enlargement was also reported in two nondiabetic models of glomerular hypertrophy. Administration of VEGF antibody in mice fed a high protein diet abolished the glomerular hypertrophy seen in placebotreated animals on an identical diet, without affecting kidney or body weight (442). Similarly, administration of VEGFantibody to uninephrectomized mice reduced the compensatory renal growth and prevented the glomerular hypertrophy (443). b. Receptor inhibitors. VEGFR tyrosine kinase inhibitors (TKi) (444) are newly designed substances, and therefore they have not yet been studied in diabetes. Currently, the effect of VEGFR-TKi on diabetic kidney disease is examined in experimental models of type 1 and type 2 diabetes. c. ACEi. The lack of association between ACE inhibition with lisinopril and circulating plasma VEGF, despite definite effects of these agents on retinopathy and nephropathy (EUCLID study), suggests that these effects are not predominantly mediated by circulating VEGF (433). In contrast, in type 1 diabetic rats, ramipril and perindopril reduced the retinal up-regulation of VEGF as measured by Northern blot analysis and in situ hybridization, respectively (445). d. AGE inhibition. In OLETF rats (319), long-term treatment with OPB-9195 conferred renoprotection and abolished the enhanced renal VEGF immunoreactivity (319).

FIG. 11. A, Albuminuria in untreated (n 12) and antiVEGF Ab-treated (n 7) control rats, and in untreated (n 11), anti-VEGF Ab-treated (n 12), and control Ab-treated (n 9) diabetic rats. *, P 0.0001 vs. control; , P 0.001 vs. diabetes; #, P 0.02 vs. diabetes control Ab; $, P 0.01 vs. control anti-VEGF Ab. B, Glomerular volume in untreated (n 6) and anti-VEGF Ab-treated (n 6) control rats, and in untreated (n 6), anti-VEGF Ab-treated (n 6), and control Ab-treated (n 6) diabetic rats. *, P 0.01 vs. control; , P 0.01 vs. diabetes; #, P 0.001 vs. diabetes control Ab. [Reproduced with permission from A. S. De Vriese et al.: J Am Soc Nephrol 12:9931000, 2001 (261). Lippincott Williams & Wilkins.]

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 995

FIG. 12. BMT and total mesangial volume on d 60 in nondiabetic controls (open bars), placebotreated diabetic db/db mice (black bars), and VEGF-antibody-treated diabetic db/db mice (gray bars). Values are means SEM (n 6 12 in each group). *, P 0.05 vs. nondiabetic controls and VEGF-antibody-treated diabetic db/db mice. [Reproduced with permission from A. Flyvbjerg et al.: Diabetes 51:3090 3094, 2002 (441). American Diabetes Association.]

e. Anti-TGF- antibodies. Anti-TGF- antibodies only slightly prevented the up-regulation of VEGF mRNA in kidneys from db/db mice (334). 7. Conclusion. Substantial studies in type 1 and type 2 diabetic animals and patients consistently demonstrate up-regulation of renal VEGF and VEGFR-2 expression, particularly early in the course of diabetes. The discrepancies in circulating VEGF levels in diabetic patients may reflect differences in sampling and assay methods or timing during the course of diabetic nephropathy. Inhibition of VEGF ameliorated the diabetesassociated renal changes in experimental models, emphasizing a deleterious role for VEGF in the pathogenesis of diabetic nephropathy. Although the cause of the VEGF upregulation in diabetes remains unknown, various factors relevant to the pathogenesis of diabetic nephropathy, including hyperglycemia, AGEs, PKC, Ang II, and TGF-1, are able to stimulate VEGF production in renal cell types. Future research will elucidate the clinical usefulness of VEGF inhibitors in the treatment of diabetic nephropathy.
D. Platelet-derived growth factor (PDGF)

and release a PDGF-like protein (449). In human kidney, PDGF-A is expressed by glomerular podocytes and tubular epithelial cells including collecting duct cells (450), whereas in mouse kidney, PDGF-A is mainly detected in epithelial cells in the loop of Henle and possibly in VSMC (451). PDGF-B is primarily expressed by human mesangial cells (449, 452), rat mesangial cells and podocytes (453, 454). In mouse kidney, however, PDGF-B is predominantly expressed by vascular endothelial cells and minimally by glomerular podocytes (451). PDGF-C is constitutively expressed by arterial smooth muscle cells and collecting duct cells in rat kidney (455). PDGF-D is expressed by glomerular podocytes and VSMCs of mature human kidney (447). In human and mouse kidney, PDGFR- is extensively expressed by interstitial cells, and occasionally by human mesangial cells and VSMCs (451, 456, 457), whereas PDGFR- is primarily expressed by mesangial cells, interstitial cells, and VSMCs (447, 451, 452, 457). In rat kidney, PDGFR- is weakly expressed by interstitial cells (451), mesangial cells, and podocytes (454). Thus, the patterns of renal expression of PDGF and its receptors differ slightly between different species. 3. In vitro evidence for PDGF effects on renal cells. Exposure to high glucose increased PDGF-B mRNA expression in cultured human mesangial cells (458) and caused changes in cell growth, collagen synthesis, and PDGF secretion in cultured human proximal tubular cells (459). Furthermore, high glucose increased PDGFR- in rat mesangial and human capillary endothelial cells through PKC activation (460). Glucose seemed to lower the threshold at which PDGF stimulates TGF-1 synthesis in cultured proximal tubular cells (461). The AGE-induced release of TGF- and production of collagens in cultured mesangial cells were mediated via PDGF (24, 462). PDGF increased RNA and protein synthesis (357) and SPARC mRNA levels (463); activated specific PKC isoforms, namely PKC, -I, -, and - in cultured mesangial cells (278); and induced mesangial cell proliferation and migration (464). 4. Experimental evidence for a role of PDGF in diabetic kidney disease. In short-term STZ-diabetic rats, PDGF-B and PDGFR- protein expression was enhanced in glomerular podocytes and mesangial cells compared with control or insulin-treated diabetic rats (454). Even very early after the onset of STZ diabetes, PDGF B-mRNA and PDGF-B immunostaining were slightly increased, preceding mesangial proliferation, increased TGF-1 expression, and ECM deposition

1. The PDGF system. The PDGF family consists of two structurally similar A- and B-polypeptide chains (PDGF-A and PDGF-B) that are able to form three disulfide-bonded homoand heterodimers, i.e., PDGF-AA, PDGF-BB, and PDGF-AB (446). PDGF-C and PDGF-D are novel growth factors belonging to the PDGF family (447, 448). Soluble PDGF binding proteins such as 2-macroglobulin, PDGF-associated protein, and the extracellular part of the PDGF- receptor as well as ECM components such as secreted protein acidic and rich in cysteine (SPARC) regulate the activity and availability of PDGF isoforms (446). PDGF is synthesized by many different cell types. PDGF isoforms exert their paracrine or autocrine effects by activating two tyrosine kinase receptors, PDGF receptor (PDGFR)- and PDGFR-, which require ligandinduced dimerization for their activation. PDGF-AA induces -receptor dimerization, PDGF-AB induces - or receptor dimerization, and PDGF-BB induces all three dimeric combinations of - and -receptors (446). PDGF has potent mitogenic and weak angiogenic effects, modulates chemotaxis, blood vessel tonus, and platelet aggregation, and is involved in tissue homeostasis (446). 2. Expression of PDGF in the normal kidney. Cultured human mesangial cells express both PDGF-A and -B chain mRNA

Downloaded from edrv.endojournals.org on April 19, 2006

996

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy

(465). In NOD mice, however, PDGF-B mRNA levels in microdissected glomeruli were unaltered (316). Long-term STZ diabetes was associated with elevated PDGF-B mRNA expression in glomeruli and tubulointerstitium (300, 466), whereas PDGF-A mRNA levels were not altered (300). Insulin treatment partially ameliorated the increase in PDGF-B mRNA in glomeruli of diabetic rats (300). In type 1 diabetic rats, a reduction in SPARC mRNA and protein has been associated with early diabetes-associated kidney growth (467). Administration of PDGF to Goto-Kakizaki rats, a model of lean type 2 diabetes, led to acute mesangial cell proliferation and activation but had no long-term effects on kidney structure or function (468). In disagreement with the in vitro findings, repeated injections of AGEs in normal mice led to an increase in the expression of type IV collagen and laminin genes in the glomeruli, and was accompanied by up-regulation of TGF-1, but not PDGF-B expression (36). 5. Clinical evidence for a role of PDGF in diabetic kidney disease. Clinical evidence for a role of PDGF in the pathogenesis of diabetic kidney disease is very limited. Serum PDGF levels did not differ between type 1 and type 2 diabetic patients without albuminuria, but the serum PDGF levels of all the diabetic patients were increased compared with agematched controls and correlated negatively with serum creatinine levels (469). Long-term type 1 diabetic patients had a higher urinary excretion of PDGF-BB than controls (470). Urinary excretion of PDGF was elevated in all diabetic subgroups, but the excretion of PDGF was higher in patients with micro- or macroalbuminuria than with normoalbuminuria, although there was a considerable overlap between the groups (470). Platelet -thromboglobulin content and PDGF were markedly decreased in 10 type 1 diabetic patients, suggesting that PDGF release might be increased in diabetic subjects (471). In a study in type 2 diabetic patients, elevated serum SPARC concentrations were detected in patients with severe tubulointerstitial lesions associated with and without advanced glomerular lesions (472). In renal biopsies from type 2 diabetic patients with overt diabetic nephropathy, PDGF-A and PDGF-B mRNA expression was up-regulated compared with control tissue (473). In addition, immunohistochemistry showed abundant expression of PDGF-A and PDGF-B in glomeruli and the tubulointerstitial compartment, whereas only minimal immunoreactive PDGF-A and PDGF-B was detected in control tissue. PDGF-A immunostaining was localized to glomerular and tubulointerstitial epithelial cells, but PDGF-B immunostaining was predominantly extracellular and localized to areas of fibrosis, suggesting that PDGF-B may be sequestered in the ECM (473). 6. Agents with effects on the PDGF system in diabetic kidney disease. Several agents inhibit the effects of PDGF by binding PDGF or by blocking the PDGFRs, i.e., oligonucleotide aptamers, low-molecular weight molecules, soluble PDGFRs, polyclonal antibodies or monoclonal PDGFR antibodies and several PDGFR-TKi. Few substances, however, have been used to investigate the role of PDGF in diabetes. Various drugs with well-known or potential therapeutic effects on diabetic kidney disease, such as ACEi, PKC inhibitors, AGE inhibitors, and ET receptor antagonists may also modulate renal PDGF expression.

a. Neutralizing antibodies. A polyclonal goat antibody, which recognizes all three dimeric forms of PDGF, abrogated the AGE-induced increase in type IV collagen mRNA in mouse mesangial cells cultured in normal glucose, suggesting that PDGF is an intermediate factor in the AGE-induced increase in ECM (24). b. PDGFR-TKi. In cultured mesangial cells, STI 571, a receptor tyrosine kinase inhibitor of the class of the 2phenylaminopyridines, reduced the PDGF-stimulated mesangial proliferation in a dose-dependent manner (474). c. ACEi. Ramipril reduced the fetal calf serum activated mesangial cell proliferation and PDGF-A and -B expression (475) and completely abolished the PKC-induced PDGF-A and -B expression, which may be independent of its ability to inhibit ACE and could represent an additional mechanism in the renal protective effects of this ACEi (475). In contrast, treatment of STZ-diabetic rats with enalapril for 24 wk reduced CrCl and urinary protein excretion, produced a nonsignificant reduction in blood pressure, but had no effect on the increased glomerular mRNA levels of PDGF-B, TNF-, TGF-, and bFGF, although it attenuated the increase in glomerular ET-1 expression (476). d. PKC inhibition. The PKC inhibitors, 1-(5-isoquinolinylsulfonyl)-2-methylpiperazine, staurosporine, and PKC inhibitor peptide, inhibited the PDGF-induced cellular proliferation and ET-1 secretion in cultured rat mesangial cells (477). e. Statins. Simvastatin inhibited PDGF-induced DNA synthesis in human glomerular mesangial cells in a dosedependent manner (478), providing evidence that statins may ameliorate glomerular pathology, at least in part, by a direct cellular effect and not only by their cholesterol-lowering potential. f. ET receptor antagonists. The mRNA levels of ECM components including 1(I), 1(III), and 1(IV) collagen; laminin B1 and B2; and growth factors including PDGF-B, TNF-, TGF-, and bFGF all increased with age in glomeruli of STZ-diabetic rats (192). Treatment for 24 wk with the specific ET receptor A antagonist FR139317 attenuated the increases in the glomerular mRNA levels of these growth factors and ECM components, attenuated the rise in CrCl, and reduced urinary protein excretion in diabetic rats, but did not affect blood pressure. g. Others. Aminoguanidine administration during 6 months attenuated the overexpression of PDGF-B mRNA in renal tubules and glomeruli of STZ-diabetic rats (466). Trapidil, an antiplatelet drug that competitively inhibits the binding of PDGF-BB to PDGFR-, inhibited PDGF-induced mesangial cell proliferation in vitro (479). In STZ-diabetic rats, daily ip injections with trapidil for 4 wk prevented glomerular hypertrophy (Fig. 13) (454). 7. Conclusion. In vitro and experimental data implicate the up-regulation of PDGF-B and PDGFR- and down-regulation of SPARC in glomeruli and tubulointerstitium in the early diabetes-associated renal hypertrophy. Furthermore,

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy

Endocrine Reviews, December 2004, 25(6):9711010 997

AGEs increase PDGF expression in vitro, but not in vivo. PDGF can stimulate ECM formation, PKC activation, and SPARC expression in cultured mesangial cells and mesangial cell proliferation both in vitro and in vivo. Diabetic patients have increased serum PDGF levels and increased urinary PDGF excretion. Overt diabetic nephropathy is associated with renal overexpression of PDGF. Trapidil, a nonspecific PDGF inhibitor, prevents glomerular hypertrophy in experimental diabetes in addition to inhibition of mesangial cell proliferation, giving further support for PDGF as a mediator of renal/glomerular growth. Future studies are warranted to explore the potential benefit of specific PDGF inhibitors in experimental diabetic kidney disease because, so far, neutralizing antibodies and PDGFR-TKi have only been investigated in cultured mesangial cells.

VI. Outlook and Future Perspectives

Tight metabolic and antihypertensive controls remain cornerstone interventions in the treatment of micro- and macrovascular complications in type 1 and type 2 diabetes. However, vascular diabetic complications, including diabetic nephropathy, remain a huge clinical problem despite implementation of overall intensified glycemic and antihypertensive control in these patients. Accordingly, there is an ongoing need for development of new therapeutic strategies in the treatment of kidney disease, which may imply that combined intervention is required toward various pathways of importance in the development of diabetic nephropathy. As described in the present review, extensive research during recent years has identified several new pathways

FIG. 13. A, Glomerular volume of control, diabetic, and insulin-treated diabetic rats. Glomerular volume in diabetic rats (closed bar) was higher than in the control rats (open bar) at all time points. Glomerular volume in diabetic rats treated with insulin (shaded bar) was lower than in the diabetic groups at 4 and 12 wk. Results were expressed as mean SD. *, P 0.05; **, P 0.01 vs. control; ##, P 0.01 vs. diabetic. C2, C4, and C12, Control rats at wk 2, 4, and 12; D2, D4, and D12, diabetic rats at wk 2, 4, and 12; DI4 and DI12, insulin-treated diabetic rats at wk 4 and 12. B, The effect of the inhibition of enhanced PDGF system on glomerular hypertrophy in diabetic rats. Glomerular volume in diabetic rats (closed bar) was higher than in the control rats (open bar), and in diabetic rats treated with trapidil (shaded bar) at 4 wk. Results were expressed as mean SD. **, P 0.01 vs. control; ##, P 0.001 vs. diabetic. C4, Control rats at 4 wk; D4, diabetic rats at 4 wk; DT4, trapidil-treated rats at 4 wk. [Reproduced with permission from H. Nakagawa et al.: Diabetes Res Clin Pract 48:8798, 2000 (454). Elsevier.]

FIG. 14. Schematic illustration of the potential hierarchy and interactions between metabolic, hemodynamic, and intracellular factors and growth factors/cytokines in the pathophysiology of diabetic kidney disease.

Downloaded from edrv.endojournals.org on April 19, 2006

998

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy sawa K, Kawata T, Koike T 1997 Progression of nephropathy in spontaneous diabetic rats is prevented by OPB-9195, a novel inhibitor of advanced glycation. Diabetes 46:895 899 Zheng F, He C, Cai W, Hattori M, Steffes M, Vlassara H 2002 Prevention of diabetic nephropathy in mice by a diet low in glycoxidation products. Diabetes Metab Res Rev 18:224 237 Wendt TM, Tanji N, Guo J, Kislinger TR, Qu W, Lu Y, Bucciarelli LG, Rong LL, Moser B, Markowitz GS, Stein G, Bierhaus A, Liliensiek B, Arnold B, Nawroth PP, Stern DM, DAgati VD, Schmidt AM 2003 RAGE drives the development of glomerulosclerosis and implicates podocyte activation in the pathogenesis of diabetic nephropathy. Am J Pathol 162:11231137 Campbell J, Davidson IWF, Lei HP 1950 The production of permanent diabetes by highly purified growth hormone. Endocrinology 46:588 590 Lundbk K, Christensen NJ, Jensen VA, Johansen K, Olsen TS, Hansen AP, rskov H, sterby R 1970 Diabetes, diabetic angiopathy, and growth hormone. Lancet 2:131133 Yde H 1969 The growth hormone dependent sulfation factor in serum from patients with various types of diabetes. Acta Med Scand 186:293297 Wendt T, Tanji N, Guo J, Hudson BI, Bierhaus A, Ramasamy R, Arnold B, Nawroth PP, Yan SF, DAgati V, Schmidt AM 2003 Glucose, glycation, and RAGE: implications for amplification of cellular dysfunction in diabetic nephropathy. J Am Soc Nephrol 14:13831395 Williams ME 2003 New therapies for advanced glycation end product nephrotoxicity: current challenges. Am J Kidney Dis 41: S42S47 Vlassara H, Palace MR 2002 Diabetes and advanced glycation endproducts. J Intern Med 251:87101 Horie K, Miyata T, Maeda K, Miyata S, Sugiyama S, Sakai H, Strihou CY, Monnier VM, Witztum JL, Kurokawa K 1997 Immunohistochemical colocalization of glycoxidation products and lipid peroxidation products in diabetic renal glomerular lesions. Implication for glycoxidative stress in the pathogenesis of diabetic nephropathy. J Clin Invest 100:29953004 Niwa T, Katsuzaki T, Miyazaki S, Miyazaki T, Ishizaki Y, Hayase F, Tatemichi N, Takei Y 1997 Immunohistochemical detection of imidazolone, a novel advanced glycation end product, in kidneys and aortas of diabetic patients. J Clin Invest 99:12721280 Tanji N, Markowitz GS, Fu C, Kislinger T, Taguchi A, Pischetsrieder M, Stern D, Schmidt AM, DAgati VD 2000 Expression of advanced glycation end products and their cellular receptor RAGE in diabetic nephropathy and nondiabetic renal disease. J Am Soc Nephrol 11:1656 1666 Soulis T, Cooper ME, Vranes D, Bucala R, Jerums G 1996 Effects of aminoguanidine in preventing experimental diabetic nephropathy are related to the duration of treatment. Kidney Int 50:627 634 Soulis T, Thallas V, Youssef S, Gilbert RE, McWilliam BG, Murray-McIntosh RP, Cooper ME 1997 Advanced glycation end products and their receptors co-localise in rat organs susceptible to diabetic microvascular injury. Diabetologia 40:619 628 Huang Y, Lin S, Zhou J 1998 Gene expression of receptor for advanced glycosylation end products and its modulation by aminoguanidine in diabetic kidney tissue. Chin Med J (Engl) 111: 698 704 He CJ, Zheng F, Stitt A, Striker L, Hattori M, Vlassara H 2000 Differential expression of renal AGE-receptor genes in NOD mice: possible role in nonobese diabetic renal disease. Kidney Int 58: 19311940 Doi T, Vlassara H, Kirstein M, Yamada Y, Striker GE, Striker LJ 1992 Receptor-specific increase in extracellular matrix production in mouse mesangial cells by advanced glycosylation end products is mediated via platelet-derived growth factor. Proc Natl Acad Sci USA 89:28732877 Skolnik EY, Yang Z, Makita Z, Radoff S, Kirstein M, Vlassara H 1991 Human and rat mesangial cell receptors for glucose-modified proteins: potential role in kidney tissue remodelling and diabetic nephropathy. J Exp Med 174:931939 Youssef S, Nguyen DT, Soulis T, Panagiotopoulos S, Jerums G, Cooper ME 1999 Effect of diabetes and aminoguanidine therapy on

with impact on the development of diabetic kidney disease. Accordingly, many potential strategies to prevent the development or retard the progression of diabetic nephropathy, independently of metabolic control and hypertension, are currently under evaluation. The challenge for future research will be to unravel these complex interactions between hyperglycemia, metabolic factors (e.g., AGE- and AR/polyol pathways), hemodynamic factors (e.g., AngII/RAS-, ET-, and NO-pathways), intracellular factors (e.g., DAG-PKC, NF-B, and MAPK), and growth factors/cytokines (e.g., TGF-, CTGF, GH, IGF, VEGF, and PDGF), which may lead to a better understanding of the pathogenesis of diabetic kidney disease. A schematic illustration of the potential hierarchy and interactions between these different systems is given in Fig. 14. The future search for new potential pathways and the development of rational strategies for improved management of diabetic nephropathy may be facilitated by the use of newly developed tools like microarray technology, glycomics, and proteomics.
Acknowledgments

9. 10.

11. 12. 13. 14.

15. Address all correspondence and requests for reprints to: Allan Flyvbjerg, M.D., D.M.Sc., Medical Department M/Medical Research Laboratories, Clinical Institute, Aarhus University Hospital, Nrrebrogade 44, DK-8000 Aarhus C, Denmark. E-mail: allan.flyvbjerg@dadlnet.dk This work was supported by the Danish Diabetes Association, the Danish Medical Research Council (Grant 9700592), the Eva and Henry Frnkels Memorial Foundation, the Nordic Insulin Foundation, the Institute of Experimental Clinical Research, University of Aarhus, Denmark, the Institute for the Promotion of Innovation by Science and Technology in Flanders (IWT), and the Fund for Scientific ResearchFlanders. Due to space limitations, many important contributions could not be referenced. 16. 17.

18.

19.

References
1. Hovind P, Tarnow L, Rossing K, Rossing P, Eising S, Larsen N, Binder C, Parving H-H 2003 Decreasing incidence of severe diabetic microangiopathy in type 1 diabetes. Diabetes Care 26:1258 1264 2. Ritz E, Keller C, Bergis K, Strojek K 1997 Pathogenesis and course of renal disease in IDDM/NIDDM: differences and similarities. Am J Hypertens 10:202S207S 3. Yokoyama H, Okudaira M, Otani T, Sato A, Miura J, Takaike H, Yamada H, Muto K, Uchigata Y, Ohashi Y, Iwamoto Y 2000 Higher incidence of diabetic nephropathy in type 2 than in type 1 diabetes in early-onset diabetes in Japan. Kidney Int 58:302311 4. Grnbk H, Vogel I, sterby R, Lancranjan I, Flyvbjerg A, rskov H 1998 Effect of octreotide, captopril or insulin on renal changes and UAE in long-term experimental diabetes. Kidney Int 53:173180 5. Noda M, Matsuo T, Nagano-Tsuge H, Ohta M, Sekiguchi M, Shibouta Y, Naka T, Imura Y 2001 Involvement of angiotensin II in progression of renal injury in rats with genetic non-insulindependent diabetes mellitus (Wistar fatty rats). Jpn J Pharmacol 85:416 422 , Yo 6. Azal O nem A, Gu ler S, C akir B, Baydar A, C orakc i A, Kutlu M 2002 Effects of aminoguanidine and tolrestat on the development of ocular and renal structural changes in experimental diabetic rats. Diabetes Obes Metab 4:7579 7. McCarthy KJ, Routh RE, Shaw W, Walsh K, Welbourne TC, Johnson JH 2000 Troglitazone halts diabetic glomerulosclerosis by blockade of mesangial expansion. Kidney Int 58:23412350 8. Nakamura S, Makita Z, Ishikawa S, Yasumura K, Fujii W, Yanagi-

20. 21.

22.

23.

24.

25.

26.

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy renal advanced glycation end-product binding. Kidney Int 55: 907916 Chen S, Cohen MP, Lautenslager GT, Shearman CW, Ziyadeh FN 2001 Glycated albumin stimulates TGF-1 production and protein kinase C activity in glomerular endothelial cells. Kidney Int 59: 673 681 Scivittaro V, Ganz MB, Weiss MF 2000 AGEs induce oxidative stress and activate protein kinase C-II in neonatal mesangial cells. Am J Physiol Renal Physiol 278:F676 F683 Cohen MP, Ziyadeh FN, Lautenslager GT, Cohen JA, Shearman CW 1999 Glycated albumin stimulation of PKC- activity is linked to increased collagen IV in mesangial cells. Am J Physiol 276:F684 F690 Ziyadeh FN, Han DC, Cohen JA, Guo J, Cohen MP 1998 Glycated albumin stimulates fibronectin gene expression in glomerular mesangial cells: involvement of the transforming growth factor- system. Kidney Int 53:631 638 Borrebk J, Prydz K, Fjeldstad K, Vuong TT, Berg TJ, Holkov C, Kolset SO 2001 The AGE product N-(carboxymethyl)lysine serum albumin is a modulator of proteoglycan expression in polarized cultured kidney epithelial cells. Diabetologia 44:488 494 Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRobert A, Thallas V, Atkins RC, Osicka T, Jerums G, Cooper ME 2001 Advanced glycation end products cause epithelial-myofibroblast transdifferentiation via the receptor for advanced glycation end products (RAGE). J Clin Invest 108:18531863 Gugliucci A, Bendayan M 1995 Reaction of advanced glycation endproducts with renal tissue from normal and streptozotocininduced diabetic rats: an ultrastructural study using colloidal gold cytochemistry. J Histochem Cytochem 43:591 600 Forbes JM, Cooper ME, Thallas V, Burns WC, Thomas MC, Brammar GC, Lee F, Grant SL, Burrell LA, Jerums G, Osicka TM 2002 Reduction of the accumulation of advanced glycation end products by ACE inhibition in experimental diabetic nephropathy. Diabetes 51:3274 3282 Soulis T, Cooper ME, Sastra S, Thallas V, Panagiotopoulos S, Bjerrum OJ, Jerums G 1997 Relative contributions of advanced glycation and nitric oxide synthase inhibition to aminoguanidinemediated renoprotection in diabetic rats. Diabetologia 40:1141 1151 Striker LJ, Striker GE 1996 Administration of AGEs in vivo induces extracellular matrix gene expression. Nephrol Dial Transplant 11(Suppl 5):62 65 Vlassara H, Fuh H, Makita Z, Krungkrai S, Cerami A, Bucala R 1992 Exogenous advanced glycosylation end products induce complex vascular dysfunction in normal animals: a model for diabetic and aging complications. Proc Natl Acad Sci USA 89:1204312047 Yamamoto Y, Kato I, Doi T, Yonekura H, Ohashi S, Takeuchi M, Watanabe T, Yamagishi S, Sakurai S, Takasawa S, Okamoto H, Yamamoto H 2001 Development and prevention of advanced diabetic nephropathy in RAGE- overexpressing mice. J Clin Invest 108:261268 Berg TJ, Clausen JT, Torjesen PA, Dahl-Jrgensen K, Bangstad HJ, Hanssen KF 1998 The advanced glycation end product N(carboxymethyl)lysine is increased in serum from children and adolescents with type 1 diabetes. Diabetes Care 21:19972002 Berg TJ, Bangstad HJ, Torjesen PA, sterby R, Bucala R, Hanssen KF 1997 Advanced glycation end products in serum predict changes in the kidney morphology of patients with insulin-dependent diabetes mellitus. Metabolism 46:661 665 He CJ, Koschinsky T, Buenting C, Vlassara H 2001 Presence of diabetic complications in type 1 diabetic patients correlates with low expression of mononuclear cell AGE-receptor-1 and elevated serum AGE. Mol Med 7:159 168 Aoki S, Hasegawa G, Shigeta H, Obayashi H, Fujii M, Kimura F, Moriwaki A, Nakamura N, Ienaga K, Nakamura K, Kondo M 2000 Crossline levels in serum and erythrocyte membrane proteins from patients with diabetic nephropathy. Diabetes Res Clin Pract 48: 119 125 Aso Y, Inukai T, Tayama K, Takemura Y 2000 Serum concentrations of advanced glycation endproducts are associated with the development of atherosclerosis as well as diabetic microangiopathy in patients with type 2 diabetes. Acta Diabetol 37:8792

Endocrine Reviews, December 2004, 25(6):9711010 999 44. Cohen MP, Sharma K, Jin Y, Hud E, Wu VY, Tomaszewski J, Ziyadeh FN 1995 Prevention of diabetic nephropathy in db/db mice with glycated albumin antagonists. A novel treatment strategy. J Clin Invest 95:2338 2345 45. Cohen MP, Clements RS, Cohen JA, Shearman CW 1996 Prevention of decline in renal function in the diabetic db/db mouse. Diabetologia 39:270 274 46. Cohen MP, Masson N, Hud E, Ziyadeh F, Han DC, Clements RS 2000 Inhibiting albumin glycation ameliorates diabetic nephropathy in the db/db mouse. Exp Nephrol 8:135143 47. Cohen MP, Ziyadeh FN, Hong SW, Shearman CW, Hud E, Lautenslager GT, Iglesias-De La Cruz MC, Chen S 2002 Inhibiting albumin glycation in vivo ameliorates glomerular overexpression of TGF-1. Kidney Int 61:20252032 48. Soulis-Liparota T, Cooper M, Papazoglou D, Clarke B, Jerums G 1991 Retardation by aminoguanidine of development of albuminuria, mesangial expansion, and tissue fluorescence in streptozocininduced diabetic rat. Diabetes 40:1328 1334 49. Osicka TM, Yu Y, Lee V, Panagiotopoulos S, Kemp BE, Jerums G 2001 Aminoguanidine and ramipril prevent diabetes-induced increases in protein kinase C activity in glomeruli, retina and mesenteric artery. Clin Sci (Colch) 100:249 257 50. Soulis T, Sastra S, Thallas V, Mortensen SB, Wilken M, Clausen JT, Bjerrum OJ, Petersen H, Lau J, Jerums G, Boel E, Cooper ME 1999 A novel inhibitor of advanced glycation end-product formation inhibits mesenteric vascular hypertrophy in experimental diabetes. Diabetologia 42:472 479 51. Osicka TM, Kiriazis Z, Pratt LM, Jerums G, Comper WD 2001 Ramipril and aminoguanidine restore renal lysosomal processing in streptozotocin diabetic rats. Diabetologia 44:230 236 52. Osicka TM, Yu Y, Panagiotopoulos S, Clavant SP, Kiriazis Z, Pike RN, Pratt LM, Russo LM, Kemp BE, Comper WD, Jerums G 2000 Prevention of albuminuria by aminoguanidine or ramipril in streptozotocin-induced diabetic rats is associated with the normalization of glomerular protein kinase C. Diabetes 49:8793 53. Wilkinson-Berka JL, Kelly DJ, Koerner SM, Jaworski K, Davis B, Thallas V, Cooper ME 2002 ALT-946 and aminoguanidine, inhibitors of advanced glycation, improve severe nephropathy in the diabetic transgenic (mREN-2)27 rat. Diabetes 51:32833289 54. Appel G, Bolton K, Freedman B, Wuerth J, Cartwright K 1999 Pimagedine (PG) lowers total urinary protein (TUP) and slows progression of overt diabetic nephropathy in patients with type 1 diabetes mellitus (DM). J Am Soc Nephrol 10:153A (Abstract) 55. Thornalley PJ 2003 Use of aminoguanidine (Pimagedine) to prevent the formation of advanced glycation endproducts. Arch Biochem Biophys 419:31 40 56. Jain SK, Lim G 2001 Pyridoxine and pyridoxamine inhibits superoxide radicals and prevents lipid peroxidation, protein glycosylation, and (Na K)-ATPase activity reduction in high glucose-treated human erythrocytes. Free Radic Biol Med 30:232237 57. Degenhardt TP, Alderson NL, Arrington DD, Beattie RJ, Basgen JM, Steffes MW, Thorpe SR, Baynes JW 2002 Pyridoxamine inhibits early renal disease and dyslipidemia in the streptozotocindiabetic rat. Kidney Int 61:939 950 58. Oturai PS, Christensen M, Rolin B, Pedersen KE, Mortensen SB, Boel E 2000 Effects of advanced glycation end-product inhibition and cross-link breakage in diabetic rats. Metabolism 49:996 1000 59. Shoda H, Miyata S, Liu BF, Yamada H, Ohara T, Suzuki K, Oimomi M, Kasuga M 1997 Inhibitory effects of tenilsetam on the Maillard reaction. Endocrinology 138:1886 1892 60. Pugliese G, Pricci F, Romeo G, Pugliese F, Mene P, Giannini S, Cresci B, Galli G, Rotella CM, Vlassara H, Di Mario U 1997 Upregulation of mesangial growth factor and extracellular matrix synthesis by advanced glycation end products via a receptor-mediated mechanism. Diabetes 46:18811887 61. Flyvbjerg A, Denner L, Schrijvers B, Tilton R, Mogensen T, Paludan SR, Rasch R 2004 Long-term renoprotective effects of a neutralizing RAGE-antibody in obese type 2 diabetic mice. Diabetes 53:166 172 62. Vasan S, Zhang X, Zhang X, Kapurniotu A, Bernhagen J, Teichberg S, Basgen J, Wagle D, Shih D, Terlecky I, Bucala R, Cerami A, Egan J, Ulrich P 1996 An agent cleaving glucose-derived protein crosslinks in vitro and in vivo. Nature 382:275278

27.

28. 29.

30.

31.

32.

33.

34.

35.

36. 37.

38.

39.

40.

41.

42.

43.

Downloaded from edrv.endojournals.org on April 19, 2006

1000

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy and protein kinase C activity in cultured mesangial cells. Diabetologia 41:362364 Kapor-Drezgic J, Zhou X, Babazono T, Dlugosz JA, Hohman T, Whiteside C 1999 Effect of high glucose on mesangial cell protein kinase C- and - is polyol pathway-dependent. J Am Soc Nephrol 10:11931203 Bleyer AJ, Fumo P, Snipes ER, Goldfarb S, Simmons DA, Ziyadeh FN 1994 Polyol pathway mediates high glucose-induced collagen synthesis in proximal tubule. Kidney Int 45:659 666 Morrisey K, Steadman R, Williams JD, Phillips AO 1999 Renal proximal tubular cell fibronectin accumulation in response to glucose is polyol pathway dependent. Kidney Int 55:160 167 Phillips AO, Steadman R, Morrisey K, Martin J, Eynstone L, Williams JD 1997 Exposure of human renal proximal tubular cells to glucose leads to accumulation of type IV collagen and fibronectin by decreased degradation. Kidney Int 52:973984 Zhang SL, Filep JG, Hohman TC, Tang SS, Ingelfinger JR, Chan JS 1999 Molecular mechanisms of glucose action on angiotensinogen gene expression in rat proximal tubular cells. Kidney Int 55: 454 464 Petrash JM, Flath M, Sens D, Bylander J 1992 Effects of osmotic stress and hyperglycemia on aldose reductase gene expression in human renal proximal tubule cells. Biochem Biophys Res Commun 187:201208 Chang WP, Dimitriadis E, Allen T, Dunlop ME, Cooper M, Larkins RG 1991 The effect of aldose reductase inhibitors on glomerular prostaglandin production and urinary albumin excretion in experimental diabetes mellitus. Diabetologia 34:225231 Faiman G, Ganguly P, Mehta A, Thliveris JA 1993 Effect of statil on kidney structure, function and polyol accumulation in diabetes mellitus. Mol Cell Biochem 125:2733 Kern TS, Engerman RL 1999 Aldose reductase and the development of renal disease in diabetic dogs. J Diabetes Complications 13:10 16 Kicic E, Palmer TN 1994 Is sorbitol dehydrogenase gene expression affected by streptozotocin-diabetes in the rat? Biochim Biophys Acta 1226:213218 Raccah D, Coste T, Cameron NE, Dufayet D, Vague P, Hohman TC 1998 Effect of the aldose reductase inhibitor tolrestat on nerve conduction velocity, Na/K ATPase activity, and polyols in red blood cells, sciatic nerve, kidney cortex, and kidney medulla of diabetic rats. J Diabetes Complications 12:154 162 Wiese TJ, Matsushita K, Lowe Jr WL, Stokes JB, Yorek MA 1996 Localization and regulation of renal Na/myo-inositol cotransporter in diabetic rats. Kidney Int 50:12021211 Raskin P, Rosenstock J 1987 Aldose reductase inhibitors and diabetic complications. Am J Med 83:298 306 Ghahary A, Chakrabarti S, Sima AA, Murphy LJ 1991 Effect of insulin and statil on aldose reductase expression in diabetic rats. Diabetes 40:13911396 Connors B, Lee WH, Wang G, Evan AP, Bohlen HG 1997 Aldose reductase and IGF-I gene expression in aortic and arteriolar smooth muscle during hypo- and hyperinsulinemic diabetes. Microvasc Res 53:53 62 Yamaoka T, Nishimura C, Yamashita K, Itakura M, Yamada T, Fujimoto J, Kokai Y 1995 Acute onset of diabetic pathological changes in transgenic mice with human aldose reductase cDNA. Diabetologia 38:255261 Shah VO, Dorin RI, Sun Y, Braun M, Zager PG 1997 Aldose reductase gene expression is increased in diabetic nephropathy. J Clin Endocrinol Metab 82:2294 2298 Moczulski DK, Scott L, Antonellis A, Rogus JJ, Rich SS, Warram JH, Krolewski AS 2000 Aldose reductase gene polymorphisms and susceptibility to diabetic nephropathy in Type 1 diabetes mellitus. Diabet Med 17:111118 Shah VO, Scavini M, Nikolic J, Sun Y, Vai S, Griffith JK, Dorin RI, Stidley C, Yacoub M, Vander Jagt DL, Eaton RP, Zager PG 1998 Z-2 microsatellite allele is linked to increased expression of the aldose reductase gene in diabetic nephropathy. J Clin Endocrinol Metab 83:2886 2891 Kouzuma T, Takahashi M, Endoh T, Kaneko R, Ura N, Shimamoto K, Watanabe N 2001 An enzymatic cycling method for the

63. Forbes JM, Soulis T, Thallas V, Panagiotopoulos S, Long DM, Vasan S, Wagle D, Jerums G, Cooper ME 2001 Renoprotective effects of a novel inhibitor of advanced glycation. Diabetologia 44:108 114 64. Hamada Y, Araki N, Horiuchi S, Hotta N 1996 Role of polyol pathway in nonenzymatic glycation. Nephrol Dial Transplant 11(Suppl 5):9598 65. Soulis-Liparota T, Cooper ME, Dunlop M, Jerums G 1995 The relative roles of advanced glycation, oxidation and aldose reductase inhibition in the development of experimental diabetic nephropathy in the Sprague-Dawley rat. Diabetologia 38:387394 66. Zheng F, Cai W, Mitsuhashi T, Vlassara H 2001 Lysozyme enhances renal excretion of advanced glycation endproducts in vivo and suppresses adverse age-mediated cellular effects in vitro: a potential AGE sequestration therapy for diabetic nephropathy? Mol Med 7:737747 67. Miyata T, Van Ypersele De Strihou C, Ueda Y, Ichimori K, Inagi R, Onogi H, Ishikawa N, Nangaku M, Kurokawa K 2002 Angiotensin II receptor antagonists and angiotensin-converting enzyme inhibitors lower in vitro the formation of advanced glycation end products: biochemical mechanisms. J Am Soc Nephrol 13:2478 2487 68. Vander Jagt DL, Robinson B, Taylor KK, Hunsaker LA 1990 Aldose reductase from human skeletal and heart muscle. Interconvertible forms related by thiol-disulfide exchange. J Biol Chem 265:2098220987 69. Narayanan S 1993 Aldose reductase and its inhibition in the control of diabetic complications. Ann Clin Lab Sci 23:148 158 70. Greene DA, Lattimer SA, Sima AA 1987 Sorbitol, phosphoinositides, and sodium-potassium-ATPase in the pathogenesis of diabetic complications. N Engl J Med 316:599 606 71. Burg MB 1995 Molecular basis of osmotic regulation. Am J Physiol 268:F983F996 72. Mallee JJ, Atta MG, Lorica V, Rim JS, Kwon HM, Lucente AD, Wang Y, Berry GT 1997 The structural organization of the human Na/myo-inositol cotransporter (SLC5A3) gene and characterization of the promoter. Genomics 46:459 465 73. Svanberg B, Ling C, Svensson PA, Johnson M, Carlsson B, Billig H 2000 Isolation of differentially expressed aldose reductase in ovaries after estrogen withdrawal from hypophysectomized diethylstilbestrol treated rats: increased expression during apoptosis. Mol Cell Endocrinol 164:183190 74. Lau ET, Cao D, Lin C, Chung SK, Chung SS 1995 Tissue-specific expression of two aldose reductase-like genes in mice: abundant expression of mouse vas deferens protein and fibroblast growth factor-regulated protein in the adrenal gland. Biochem J 312(Pt 2):609 615 75. Robinson B, Hunsaker LA, Stangebye LA, Vander Jagt DL 1993 Aldose and aldehyde reductases from human kidney cortex and medulla. Biochim Biophys Acta 1203:260 266 76. Burger-Kentischer A, Muller E, Neuhofer W, Marz J, Thurau K, Beck F 1999 Expression of aldose reductase, sorbitol dehydrogenase and Na/myo-inositol and Na/Cl/betaine transporter mRNAs in individual cells of the kidney during changes in the diuretic state. Pflugers Arch 437:248 254 77. Kasajima H, Yamagishi S, Sugai S, Yagihashi N, Yagihashi S 2001 Enhanced in situ expression of aldose reductase in peripheral nerve and renal glomeruli in diabetic patients. Virchows Arch 439:46 54 78. Ludvigson MA, Sorenson RL 1980 Immunohistochemical localization of aldose reductase. II. Rat eye and kidney. Diabetes 29: 450 459 79. Kikkawa R, Umemura K, Haneda M, Arimura T, Ebata K, Shigeta Y 1987 Evidence for existence of polyol pathway in cultured rat mesangial cells. Diabetes 36:240 243 80. Williamson JR, Chang K, Frangos M, Hasan KS, Ido Y, Kawamura T, Nyengaard JR, van den Enden M, Kilo C, Tilton RG 1993 Hyperglycemic pseudohypoxia and diabetic complications. Diabetes 42:801 813 81. Henry DN, Busik JV, Brosius III FC, Heilig CW 1999 Glucose transporters control gene expression of aldose reductase, PKC, and GLUT1 in mesangial cells in vitro. Am J Physiol 277:F97F104 82. Ishii H, Tada H, Isogai S 1998 An aldose reductase inhibitor prevents glucose-induced increase in transforming growth factor-

83.

84. 85. 86.

87.

88.

89.

90. 91. 92. 93.

94. 95. 96. 97.

98.

99. 100.

101.

102.

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy measurement of myo-inositol in biological samples. Clin Chim Acta 312:143151 Yoshii H, Uchino H, Ohmura C, Watanabe K, Tanaka Y, Kawamori R 2001 Clinical usefulness of measuring urinary polyol excretion by gas-chromatography/mass-spectrometry in type 2 diabetes to assess polyol pathway activity. Diabetes Res Clin Pract 51:115123 Costantino L, Rastelli G, Vianello P, Cignarella G, Barlocco D 1999 Diabetes complications and their potential prevention: aldose reductase inhibition and other approaches. Med Res Rev 19:323 Isogai S, Inokuchi T, Ohe K 1995 Effect of an aldose reductase inhibitor on glomerular basement membrane anionic sites in streptozotocin-induced diabetic rats. Diabetes Res Clin Pract 30:111116 Ishii N, Ikenaga H, Ogawa Z, Aoki Y, Saruta T, Suga T 2001 Effects of renal sorbitol accumulation on urinary excretion of enzymes in hyperglycaemic rats. Ann Clin Biochem 38:391398 Itagaki I, Shimizu K, Kamanaka Y, Ebata K, Kikkawa R, Haneda M, Shigeta Y 1994 The effect of an aldose reductase inhibitor (Epalrestat) on diabetic nephropathy in rats. Diabetes Res Clin Pract 25:147154 Pedersen MM, Christiansen JS, Mogensen CE 1991 Reduction of glomerular hyperfiltration in normoalbuminuric IDDM patients by 6 mo of aldose reductase inhibition. Diabetes 40:527531 Passariello N, Sepe J, Marrazzo G, De Cicco A, Peluso A, Pisano MC, Sgambato S, Tesauro P, DOnofrio F 1993 Effect of aldose reductase inhibitor (tolrestat) on urinary albumin excretion rate and glomerular filtration rate in IDDM subjects with nephropathy. Diabetes Care 16:789 795 Ranganathan S, Krempf M, Feraille E, Charbonnel B 1993 Short term effect of an aldose reductase inhibitor on urinary albumin excretion rate (UAER) and glomerular filtration rate (GFR) in type 1 diabetic patients with incipient nephropathy. Diabete Metab 19: 257261 Iso K, Tada H, Kuboki K, Inokuchi T 2001 Long-term effect of epalrestat, an aldose reductase inhibitor, on the development of incipient diabetic nephropathy in Type 2 diabetic patients. J Diabetes Complications 15:241244 Rastelli G, Ferrari AM, Costantino L, Gamberini MC 2002 Discovery of new inhibitors of aldose reductase from molecular docking and database screening. Bioorg Med Chem 10:14371450 Carey RM, Siragy HM 2003 Newly recognized components of the renin-angiotensin system: potential roles in cardiovascular and renal regulation. Endocr Rev 24:261271 Cao Z, Kelly DJ, Cox A, Casley D, Forbes JM, Martinello P, Dean R, Gilbert RE, Cooper ME 2000 Angiotensin type 2 receptor is expressed in the adult rat kidney and promotes cellular proliferation and apoptosis. Kidney Int 58:24372451 Lai KN, Leung JC, Lai KB, To WY, Yeung VT, Lai FM 1998 Gene expression of the renin-angiotensin system in human kidney. J Hypertens 16:91102 Zimpelmann J, Kumar D, Levine DZ, Wehbi G, Imig JD, Navar LG, Burns KD 2000 Early diabetes mellitus stimulates proximal tubule renin mRNA expression in the rat. Kidney Int 58:2320 2330 Miyata N, Park F, Li XF, Cowley Jr AW 1999 Distribution of angiotensin AT1 and AT2 receptor subtypes in the rat kidney. Am J Physiol 277:F437F446 Ozono R, Wang ZQ, Moore AF, Inagami T, Siragy HM, Carey RM 1997 Expression of the subtype 2 angiotensin (AT2) receptor protein in rat kidney. Hypertension 30:1238 1246 Wehbi GJ, Zimpelmann J, Carey RM, Levine DZ, Burns KD 2001 Early streptozotocin-diabetes mellitus downregulates rat kidney AT2 receptors. Am J Physiol Renal Physiol 280:F254 F265 Chouinard RF, Meek RL, Cooney SK, Tuttle KR 2002 Effects of amino acids and glucose on mesangial cell aminopeptidase A and angiotensin receptors. Kidney Int 61(Suppl 1):106 109 Singh R, Alavi N, Singh AK, Leehey DJ 1999 Role of angiotensin II in glucose-induced inhibition of mesangial matrix degradation. Diabetes 48:2066 2073 Ikeda M, Kohno M, Takeda T 1995 Endothelin production in cultured mesangial cells of spontaneously hypertensive rats. Hypertension 25:1196 1201 Bakris GL, Re RN 1993 Endothelin modulates angiotensin II-

Endocrine Reviews, December 2004, 25(6):9711010 1001 induced mitogenesis of human mesangial cells. Am J Physiol 264: F937F942 Ruperez M, Ruiz-Ortega M, Esteban V, Lorenzo O, Mezzano S, Plaza JJ, Egido J 2003 Angiotensin II increases connective tissue growth factor in the kidney. Am J Pathol 163:19371947 Bhaskaran M, Reddy K, Radhakrishanan N, Franki N, Ding G, Singhal PC 2003 Angiotensin II induces apoptosis in renal proximal tubular cells. Am J Physiol Renal Physiol 284:F955F965 Burns KD 2000 Angiotensin II and its receptors in the diabetic kidney. Am J Kidney Dis 36:449 467 Kelly DJ, Skinner SL, Gilbert RE, Cox AJ, Cooper ME, WilkinsonBerka JL 2000 Effects of endothelin or angiotensin II receptor blockade on diabetes in the transgenic (mRen-2)27 rat. Kidney Int 57: 18821894 Mizuiri S, Yoshikawa H, Tanegashima M, Miyagi M, Kobayashi M, Sakai K, Hayashi I, Aikawa A, Ohara T, Hasegawa A 1998 Renal ACE immunohistochemical localization in NIDDM patients with nephropathy. Am J Kidney Dis 31:301307 Wagner J, Gehlen F, Ciechanowicz A, Ritz E 1999 Angiotensin II receptor type 1 gene expression in human glomerulonephritis and diabetes mellitus. J Am Soc Nephrol 10:545551 Fogarty DG, Harron JC, Hughes AE, Nevin NC, Doherty CC, Maxwell AP 1996 A molecular variant of angiotensinogen is associated with diabetic nephropathy in IDDM. Diabetes 45:1204 1208 Schmidt S, Giessel R, Bergis KH, Strojek K, Grzeszczak W, Ganten D, Ritz E 1996 Angiotensinogen gene M235T polymorphism is not associated with diabetic nephropathy. The Diabetic Nephropathy Study Group. Nephrol Dial Transplant 11:17551761 Tarnow L, Kjeld T, Knudsen E, Major-Pedersen A, Parving H-H 2000 Lack of synergism between long-term poor glycaemic control and three gene polymorphisms of the renin angiotensin system on risk of developing diabetic nephropathy in type I diabetic patients. Diabetologia 43:794 799 Wong TY, Chan JC, Poon E, Li PK 1999 Lack of association of angiotensin-converting enzyme (DD/II) and angiotensinogen M235T gene polymorphism with renal function among Chinese patients with type II diabetes. Am J Kidney Dis 33:1064 1070 Rudberg S, Rasmussen LM, Bangstad HJ, sterby R 2000 Influence of insertion/deletion polymorphism in the ACE-I gene on the progression of diabetic glomerulopathy in type 1 diabetic patients with microalbuminuria. Diabetes Care 23:544 548 Tarnow L, Gluud C, Parving H-H 1998 Diabetic nephropathy and the insertion/deletion polymorphism of the angiotensin-converting enzyme gene. Nephrol Dial Transplant 13:11251130 Taal MW, Brenner BM 2002 Combination ACEI and ARB therapy: additional benefit in renoprotection? Curr Opin Nephrol Hypertens 11:377381 Birkenhager WH, de Leeuw PW 1999 Non-peptide angiotensin type 1 receptor antagonists in the treatment of hypertension. J Hypertens 17:873 881 Weir MR, Henrich WL 2000 Theoretical basis and clinical evidence for differential effects of angiotensin-converting enzyme inhibitors and angiotensin II receptor subtype 1 blockers. Curr Opin Nephrol Hypertens 9:403 411 Remuzzi G, Ruggenenti P, Benigni A 1997 Understanding the nature of renal disease progression. Kidney Int 51:215 Nankervis A, Nicholls K, Kilmartin G, Allen P, Ratnaike S, Martin FI 1998 Effects of perindopril on renal histomorphometry in diabetic subjects with microalbuminuria: a 3-year placebocontrolled biopsy study. Metabolism 47:1215 sterby R, Bangstad HJ, Rudberg S 2000 Follow-up study of glomerular dimensions and cortical interstitium in microalbuminuric type 1 diabetic patients with or without antihypertensive treatment. Nephrol Dial Transplant 15:1609 1616 Rudberg S, sterby R, Bangstad HJ, Dahlquist G, Persson B 1999 Effect of angiotensin converting enzyme inhibitor or blocker on glomerular structural changes in young microalbuminuric patients with type I (insulin-dependent) diabetes mellitus. Diabetologia 42:589 595 2001 Effect of 3 years of antihypertensive therapy on renal structure in type 1 diabetic patients with albuminuria: the European Study

103.

124. 125. 126. 127.

104. 105. 106. 107.

128.

129. 130.

108. 109.

131.

110.

132.

111.

133.

112. 113. 114.

134.

135. 136. 137. 138.

115. 116. 117. 118. 119. 120. 121. 122. 123.

139. 140.

141.

142.

143.

Downloaded from edrv.endojournals.org on April 19, 2006

1002

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 345:861 869 Richer C, Bruneval P, Menard J, Giudicelli JF 1998 Additive effects of enalapril and losartan in (mREN-2)27 transgenic rats. Hypertension 31:692 698 Wilkinson-Berka JL, Gibbs NJ, Cooper ME, Skinner SL, Kelly DJ 2001 Renoprotective and anti-hypertensive effects of combined valsartan and perindopril in progressive diabetic nephropathy in the transgenic (mRen-2)27 rat. Nephrol Dial Transplant 16:1343 1349 Cao Z, Bonnet F, Davis B, Allen TJ, Cooper ME 2001 Additive hypotensive and anti-albuminuric effects of angiotensin-converting enzyme inhibition and angiotensin receptor antagonism in diabetic spontaneously hypertensive rats. Clin Sci (Lond) 100: 591599 Jacobsen P, Andersen S, Rossing K, Hansen BV, Parving H-H 2002 Dual blockade of the renin-angiotensin system in type 1 patients with diabetic nephropathy. Nephrol Dial Transplant 17:1019 1024 Rossing K, Christensen PK, Jensen BR, Parving H-H 2002 Dual blockade of the renin-angiotensin system in diabetic nephropathy: a randomized double-blind crossover study. Diabetes Care 25: 95100 Mogensen CE, Neldam S, Tikkanen I, Oren S, Viskoper R, Watts RW, Cooper ME 2000 Randomised controlled trial of dual blockade of renin-angiotensin system in patients with hypertension, microalbuminuria, and non-insulin dependent diabetes: the candesartan and lisinopril microalbuminuria (CALM) study. BMJ 321: 1440 1444 Abassi ZA, Ellahham S, Winaver J, Hoffman A 2001 The intrarenal endothelin system and hypertension. News Physiol Sci 16:152156 Naicker S, Bhoola KD 2001 Endothelins: vasoactive modulators of renal function in health and disease. Pharmacol Ther 90:61 88 Kedzierski RM, Yanagisawa M 2001 Endothelin system: the double-edged sword in health and disease. Annu Rev Pharmacol Toxicol 41:851 876 Pupilli C, Romagnani P, Lasagni L, Bellini F, Misciglia N, Emoto N, Yanagisawa M, Rizzo M, Mannelli M, Serio M 1997 Localization of endothelin-converting enzyme-1 in human kidney. Am J Physiol 273:F749 F756 Kohan DE 1992 Production of endothelin-1 by rat mesangial cells: regulation by tumor necrosis factor. J Lab Clin Med 119:477 484 Sakamoto H, Sasaki S, Nakamura Y, Fushimi K, Marumo F 1992 Regulation of endothelin-1 production in cultured rat mesangial cells. Kidney Int 41:350 355 Zoja C, Orisio S, Perico N, Benigni A, Morigi M, Benatti L, Rambaldi A, Remuzzi G 1991 Constitutive expression of endothelin gene in cultured human mesangial cells and its modulation by transforming growth factor-, thrombin, and a thromboxane A2 analogue. Lab Invest 64:16 20 Kasinath BS, Fried TA, Davalath S, Marsden PA 1992 Glomerular epithelial cells synthesize endothelin peptides. Am J Pathol 141: 279 283 Karet FE, Davenport AP 1996 Localization of endothelin peptides in human kidney. Kidney Int 49:382387 Zoja C, Morigi M, Figliuzzi M, Bruzzi I, Oldroyd S, Benigni A, Ronco P, Remuzzi G 1995 Proximal tubular cell synthesis and secretion of endothelin-1 on challenge with albumin and other proteins. Am J Kidney Dis 26:934 941 Kitamura K, Tanaka T, Kato J, Eto T, Tanaka K 1989 Regional distribution of immunoreactive endothelin in porcine tissue: abundance in inner medulla of kidney. Biochem Biophys Res Commun 161:348 352 Uchida S, Takemoto F, Ogata E, Kurokawa K 1992 Detection of endothelin-1 mRNA by RT-PCR in isolated rat renal tubules. Biochem Biophys Res Commun 188:108 113 Terada Y, Tomita K, Nonoguchi H, Yang T, Marumo F 1993 Expression of endothelin-3 mRNA along rat nephron segments using polymerase chain reaction. Kidney Int 44:12731280 Sorokin A, Kohan DE 2003 Physiology and pathology of endothelin-1 in renal mesangium. Am J Physiol Renal Physiol 285:F579 F589 Fukui M, Nakamura T, Ebihara I, Osada S, Tomino Y, Masaki T,

144.

145.

146.

147.

148.

149.

150. 151. 152.

153. 154.

155.

156. 157.

158. 159.

160.

161.

for the Prevention of Renal Disease in Type 1 Diabetes (ESPRIT) Study Group. Diabetes 50:843 850 Pedersen MM, Schmitz A, Pedersen EB, Danielsen H, Christiansen JS 1988 Acute and long-term renal effects of angiotensin converting enzyme inhibition in normotensive, normoalbuminuric insulin-dependent diabetic patients. Diabet Med 5:562569 de Azevedo MJ, Ramos OL, Gross JL 1997 Lack of effect of captopril on glomerular hyperfiltration in normoalbuminuric normotensive insulin-dependent diabetic patients. Horm Metab Res 29: 516 519 OHare P, Bilbous R, Mitchell T, OCallaghan CJ, Viberti GC 2000 Low-dose ramipril reduces microalbuminuria in type 1 diabetic patients without hypertension: results of a randomized controlled trial. Diabetes Care 23:18231829 Jerums G, Allen TJ, Campbell DJ, Cooper ME, Gilbert RE, Hammond JJ, Raffaele J, Tsalamandris C 2001 Long-term comparison between perindopril and nifedipine in normotensive patients with type 1 diabetes and microalbuminuria. Am J Kidney Dis 37: 890 899 Pedersen MM, Christensen CK, Hansen KW, Christiansen JS, Mogensen CE 1991 ACE-inhibition and renoprotection in early diabetic nephropathy. Response to enalapril acutely and in longterm combination with conventional antihypertensive treatment. Clin Invest Med 14:642 651 Ciavarella A, Mustacchio A, Silletti A, Franchi R, Levorato M, Campieri C, Borgnino LC, Capozzi G, Morotti L, Vannini P 1992 Low-dose angiotensin converting enzyme inhibitors: effect on renal function in normo- and hypertensive type 1 diabetic patients. Eur J Med 1:268 272 Parving H-H, Hommel E, Jensen BR, Hansen HP 2001 Long-term beneficial effect of ACE inhibition on diabetic nephropathy in normotensive type 1 diabetic patients. Kidney Int 60:228 234 Lewis EJ, Hunsicker LG, Bain RP, Rohde RD 1993 The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med 329:1456 1462 Holdaas H, Hartmann A, Berg KJ, Langberg H, Blystad L, Fauchald P 1995 Contrasting effects of angiotensin converting inhibitor and -1-antagonist on albuminuria in insulin-dependent diabetes mellitus patients with nephropathy. J Intern Med 237:6371 Nakamura T, Ushiyama C, Suzuki S, Hara M, Shimada N, Ebihara I, Koide H 2000 Urinary excretion of podocytes in patients with diabetic nephropathy. Nephrol Dial Transplant 15:1379 1383 Ruggenenti P, Mosconi L, Sangalli F, Casiraghi F, Gambara V, Remuzzi G, Remuzzi A 1999 Glomerular size-selective dysfunction in NIDDM is not ameliorated by ACE inhibition or by calcium channel blockade. Kidney Int 55:984 994 Ravid M, Brosh D, Levi Z, Bar-Dayan Y, Ravid D, Rachmani R 1998 Use of enalapril to attenuate decline in renal function in normotensive, normoalbuminuric patients with type 2 diabetes mellitus. A randomized, controlled trial. Ann Intern Med 128: 982988 Ravid M, Savin H, Jutrin I, Bental T, Lang R, Lishner M 1994 Long-term effect of ACE inhibition on development of nephropathy in diabetes mellitus type II. Kidney Int Suppl 45:S161S164 Chan JC, Ko GT, Leung DH, Cheung RC, Cheung MY, So WY, Swaminathan R, Nicholls MG, Critchley JA, Cockram CS 2000 Long-term effects of angiotensin-converting enzyme inhibition and metabolic control in hypertensive type 2 diabetic patients. Kidney Int 57:590 600 Mizuno M, Sada T, Kato M, Koike H 2002 Renoprotective effects of blockade of angiotensin II AT1 receptors in an animal model of type 2 diabetes. Hypertens Res 25:271278 Parving H-H, Lehnert H, Brochner-Mortensen J, Gomis R, Andersen S, Arner P 2001 The effect of irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. N Engl J Med 345:870 878 Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, Ritz E, Atkins RC, Rohde R, Raz I 2001 Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med 345:851 860 Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S 2001

162. 163.

164.

165.

166.

167.

168. 169. 170. 171.

172. 173. 174.

175. 176. 177.

178.

179. 180. 181. 182.

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy Goto K, Furuichi Y, Koide H 1993 Gene expression for endothelins and their receptors in glomeruli of diabetic rats. J Lab Clin Med 122:149 156 Karet FE, Davenport AP 1995 Comparative quantification of endothelin receptor mRNA in human kidney: new tools for direct investigation of human tissue. J Cardiovasc Pharmacol 26(Suppl 3):S268 S271 Marsden PA, Dorfman DM, Collins T, Brenner BM, Orkin SH, Ballermann BJ 1991 Regulated expression of endothelin 1 in glomerular capillary endothelial cells. Am J Physiol 261:F117F125 Schnermann JB, Zhu XL, Shu X, Yang T, Huang YG, Kretzler M, Briggs JP 1996 Regulation of endothelin production and secretion in cultured collecting duct cells by endogenous transforming growth factor-. Endocrinology 137:5000 5008 Hocher B, Thone-Reineke C, Bauer C, Raschack M, Neumayer HH 1997 The paracrine endothelin system: pathophysiology and implications in clinical medicine. Eur J Clin Chem Clin Biochem 35:175189 Hocher B, Lun A, Priem F, Neumayer HH, Raschack M 1998 Renal endothelin system in diabetes: comparison of angiotensin- converting enzyme inhibition and endothelin-A antagonism. J Cardiovasc Pharmacol 31(Suppl 1):S492S495 Deuther-Conrad W, Franke S, Henle T, Sommer M, Stein G 2001 In vitro-prepared advanced glycation end-products and the modulating potential of their low-molecular weight degradation products in IRPTC-A rat proximal-tubular derived kidney epithelial cell line. Cell Mol Biol (Noisy -le-grand) 47 Online Pub:OL187OL196 Glogowski EA, Tsiani E, Zhou X, Fantus IG, Whiteside C 1999 High glucose alters the response of mesangial cell protein kinase C isoforms to endothelin-1. Kidney Int 55:486 499 Takahashi K, Suda K, Lam HC, Ghatei MA, Bloom SR 1991 Endothelin-like immunoreactivity in rat models of diabetes mellitus. J Endocrinol 130:123127 Shin SJ, Lee YJ, Lin SR, Tan MS, Lai YH, Tsai JH 1995 Decrease of renal endothelin 1 content and gene expression in diabetic rats with moderate hyperglycemia. Nephron 70:486 493 Nakamura T, Ebihara I, Fukui M, Tomino Y, Koide H 1995 Effect of a specific endothelin receptor A antagonist on mRNA levels for extracellular matrix components and growth factors in diabetic glomeruli. Diabetes 44:895 899 Hopfner RL, Misurski D, Wilson TW, McNeill JR, Gopalakrishnan V 1998 Insulin and vanadate restore decreased plasma endothelin concentrations and exaggerated vascular responses to normal in the streptozotocin diabetic rat. Diabetologia 41:12331240 Awazu M, Parker RE, Harvie BR, Ichikawa I, Kon V 1991 Downregulation of endothelin-1 receptors by protein kinase C in streptozotocin diabetic rats. J Cardiovasc Pharmacol 17(Suppl 7):S500 S502 Gross ML, Ritz E, Schoof A, Helmke B, Parkman A, Tulp O, Munter K, Amann K 2003 Renal damage in the SHR/N-cp type 2 diabetes model: comparison of an angiotensin-converting enzyme inhibitor and endothelin receptor blocker. Lab Invest 83:12671277 Hocher B, Thone-Reineke C, Rohmeiss P, Schmager F, Slowinski T, Burst V, Siegmund F, Quertermous T, Bauer C, Neumayer HH, Schleuning WD, Theuring F 1997 Endothelin-1 transgenic mice develop glomerulosclerosis, interstitial fibrosis, and renal cysts but not hypertension. J Clin Invest 99:1380 1389 Hocher B, Liefeldt L, Thone-Reineke C, Orzechowski HD, Distler A, Bauer C, Paul M 1996 Characterization of the renal phenotype of transgenic rats expressing the human endothelin-2 gene. Hypertension 28:196 201 Ciarla MV, Bocciarelli A, Di Gregorio S, Tordi A, Cotroneo P, Marra G, Ghirlanda G, Strom R 2001 Autoantibodies and endothelial dysfunction in well-controlled, uncomplicated insulindependent diabetes mellitus patients. Atherosclerosis 158:241246 Neri S, Bruno CM, Leotta C, DAmico RA, Pennisi G, Ierna D 1998 Early endothelial alterations in non-insulin-dependent diabetes mellitus. Int J Clin Lab Res 28:100 103 Iwase M, Doi Y, Goto D, Ichikawa K, Iino K, Yoshinari M, Fujishima M 2000 Effect of nicardipine versus enalapril on plasma endothelin-1 in hypertensive patients with type 2 diabetes mellitus. Clin Exp Hypertens 22:695703 Schneider JG, Tilly N, Hierl T, Sommer U, Hamann A, Dugi K,

Endocrine Reviews, December 2004, 25(6):9711010 1003 Leidig-Bruckner G, Kasperk C 2002 Elevated plasma endothelin-1 levels in diabetes mellitus. Am J Hypertens 15:967972 Shin SJ, Lee YJ, Tsai JH 1996 The correlation of plasma and urine endothelin-1 with the severity of nephropathy in Chinese patients with type 2 diabetes. Scand J Clin Lab Invest 56:571576 Totsune K, Sone M, Takahashi K, Ohneda M, Itoi K, Murakami O, Saito T, Mouri T, Yoshinaga K 1991 Immunoreactive endothelin in urine of patients with and without diabetes mellitus. J Cardiovasc Pharmacol 17(Suppl 7):S423S424 Yao J, Morioka T, Li B, Oite T 2001 Endothelin is a potent inhibitor of matrix metalloproteinase-2 secretion and activation in rat mesangial cells. Am J Physiol Renal Physiol 280:F628 F635 Li M, Chen Y, Zhang Z, Gao J 1999 Inhibition of mesangial cell proliferation by antisense oligodeoxynucleotide targeting preproendothelin-1 mRNA in vitro. Chin Med J (Engl) 112:790 793 Benigni A, Colosio V, Brena C, Bruzzi I, Bertani T, Remuzzi G 1998 Unselective inhibition of endothelin receptors reduces renal dysfunction in experimental diabetes. Diabetes 47:450 456 Nakamura T, Ebihara I, Tomino Y, Koide H 1996 Alteration of growth-related proto-oncogene expression in diabetic glomeruli by a specific endothelin receptor A antagonist. Nephrol Dial Transplant 11:1528 1531 Barton M, Shaw S, dUscio LV, Moreau P, Lu scher TF 1997 Angiotensin II increases vascular and renal endothelin-1 and functional endothelin converting enzyme activity in vivo: role of ETA receptors for endothelin regulation. Biochem Biophys Res Commun 238:861 865 Dhein S, Hochreuther S, Aus Dem Spring C, Bollig K, Hufnagel C, Raschack M 2000 Long-term effects of the endothelinA receptor antagonist LU 135252 and the angiotensin-converting enzyme inhibitor trandolapril on diabetic angiopathy and nephropathy in a chronic type I diabetes mellitus rat model. J Pharmacol Exp Ther 293:351359 Hocher B, Schwarz A, Reinbacher D, Jacobi J, Lun A, Priem F, Bauer C, Neumayer HH, Raschack M 2001 Effects of endothelin receptor antagonists on the progression of diabetic nephropathy. Nephron 87:161169 Komers R, Anderson S 2003 Paradoxes of nitric oxide in the diabetic kidney. Am J Physiol Renal Physiol 284:F1121F1137 Alderton WK, Cooper CE, Knowles RG 2001 Nitric oxide synthases: structure, function and inhibition. Biochem J 357:593 615 Kone BC, Baylis C 1997 Biosynthesis and homeostatic roles of nitric oxide in the normal kidney. Am J Physiol 272:F561F578 Vallance P, Leone A, Calver A, Collier J, Moncada S 1992 Accumulation of an endogenous inhibitor of nitric oxide synthesis in chronic renal failure. Lancet 339:572575 Lin KY, Ito A, Asagami T, Tsao PS, Adimoolam S, Kimoto M, Tsuji H, Reaven GM, Cooke JP 2002 Impaired nitric oxide synthase pathway in diabetes mellitus: role of asymmetric dimethylarginine and dimethylarginine dimethylaminohydrolase. Circulation 106:987992 Kone BC 1999 Localization and regulation of nitric oxide synthase isoforms in the kidney. Semin Nephrol 19:230 241 Bachmann S, Bosse HM, Mundel P 1995 Topography of nitric oxide synthesis by localizing constitutive NO synthases in mammalian kidney. Am J Physiol 268:F885F898 Komers R, Lindsley JN, Oyama TT, Allison KM, Anderson S 2000 Role of neuronal nitric oxide synthase (NOS1) in the pathogenesis of renal hemodynamic changes in diabetes. Am J Physiol Renal Physiol 279:F573F583 Furusu A, Miyazaki M, Abe K, Tsukasaki S, Shioshita K, Sasaki O, Miyazaki K, Ozono Y, Koji T, Harada T, Sakai H, Kohno S 1998 Expression of endothelial and inducible nitric oxide synthase in human glomerulonephritis. Kidney Int 53:1760 1768 Goto S, Yamamoto T, Feng L, Yaoita E, Hirose S, Fujinaka H, Kawasaki K, Hattori R, Yui Y, Wilson CB 1995 Expression and localization of inducible nitric oxide synthase in anti-Thy-1 glomerulonephritis. Am J Pathol 147:11331141 Brodsky SV, Morrishow AM, Dharia N, Gross SS, Goligorsky MS 2001 Glucose scavenging of nitric oxide. Am J Physiol Renal Physiol 280:F480 F486 Prabhakar SS 2001 Tetrahydrobiopterin reverses the inhibition of

202. 203.

183.

184. 185.

204. 205. 206. 207.

186.

187.

188.

208.

189. 190. 191. 192.

209.

210.

211. 212. 213. 214. 215.

193.

194.

195.

196.

216. 217. 218.

197.

198.

219.

199. 200.

220.

221. 222.

201.

Downloaded from edrv.endojournals.org on April 19, 2006

1004

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy attributed to endothelium-derived hyperpolarizing factor in streptozotocin-induced diabetic rats is restored by 5-methyltetrahydrofolate. Diabetologia 43:1116 1125 OByrne S, Forte P, Roberts LJ, Morrow JD, Johnston A, Anggard E, Leslie RD, Benjamin N 2000 Nitric oxide synthesis and isoprostane production in subjects with type 1 diabetes and normal urinary albumin excretion. Diabetes 49:857 862 Chiarelli F, Cipollone F, Romano F, Tumini S, Costantini F, di Ricco L, Pomilio M, Pierdomenico SD, Marini M, Cuccurullo F, Mezzetti A 2000 Increased circulating nitric oxide in young patients with type 1 diabetes and persistent microalbuminuria: relation to glomerular hyperfiltration. Diabetes 49:1258 1263 Maejima K, Nakano S, Himeno M, Tsuda S, Makiishi H, Ito T, Nakagawa A, Kigoshi T, Ishibashi T, Nishio M, Uchida K 2001 Increased basal levels of plasma nitric oxide in type 2 diabetic subjects. Relationship to microvascular complications. J Diabetes Complications 15:135143 Daimon M, Sugiyama K, Saitoh T, Yamaguchi H, Hirata A, Ohnuma H, Igarashi M, Eguchi H, Manaka H, Kato T 2000 Increase in serum ceruloplasmin levels is correlated with a decrease of serum nitric oxide levels in type 2 diabetes. Diabetes Care 23: 559 560 Hiragushi K, Sugimoto H, Shikata K, Yamashita T, Miyatake N, Shikata Y, Wada J, Kumagai I, Fukushima M, Makino H 2001 Nitric oxide system is involved in glomerular hyperfiltration in Japanese normo- and micro-albuminuric patients with type 2 diabetes. Diabetes Res Clin Pract 53:149 159 Francesconi F, Mingardi R, deKreutzenberg S, Avogaro A 2001 Effect of metabolic control on nitrite and nitrate metabolism in type 2 diabetic patients. Clin Exp Pharmacol Physiol 28:518 521 Miyazaki H, Matsuoka H, Cooke JP, Usui M, Ueda S, Okuda S, Imaizumi T 1999 Endogenous nitric oxide synthase inhibitor: a novel marker of atherosclerosis. Circulation 99:11411146 Abbasi F, Asagmi T, Cooke JP, Lamendola C, McLaughlin T, Reaven GM, Stuehlinger M, Tsao PS 2001 Plasma concentrations of asymmetric dimethylarginine are increased in patients with type 2 diabetes mellitus. Am J Cardiol 88:12011203 Ito A, Egashira K, Narishige T, Muramatsu K, Takeshita A 2002 Angiotensin-converting enzyme activity is involved in the mechanism of increased endogenous nitric oxide synthase inhibitor in patients with type 2 diabetes mellitus. Circ J 66:811 815 Asagami T, Abbasi F, Stuelinger M, Lamendola C, McLaughlin T, Cooke JP, Reaven GM, Tsao PS 2002 Metformin treatment lowers asymmetric dimethylarginine concentrations in patients with type 2 diabetes. Metabolism 51:843 846 Mattar AL, Fujihara CK, Ribeiro MO, de Nucci G, Zatz R 1996 Renal effects of acute and chronic nitric oxide inhibition in experimental diabetes. Nephron 74:136 143 Shultz PJ, Brown DM, Mauer SM 1995 Diabetic rats have blunted vasoconstrictive responses to chronic nitric oxide (NO) inhibition. J Am Soc Nephrol 6:1048 (Abstract) Kiff RJ, Gardiner SM, Compton AM, Bennett T 1991 The effects of endothelin-1 and NG-nitro-l-arginine methyl ester on regional haemodynamics in conscious rats with streptozotocin-induced diabetes mellitus. Br J Pharmacol 103:13211326 Ohishi K, Carmines PK 1995 Superoxide dismutase restores the influence of nitric oxide on renal arterioles in diabetes mellitus. J Am Soc Nephrol 5:1559 1566 Dalla Vestra M, Sacerdoti D, Bombonato G, Fioretto P, Finucci G, Saller A, Sfriso A, Bruseghin M, Sambataro M, Velussi M, Baggio B, Nosadini R, Crepaldi G 2002 Nitric oxide modulation of renal and cardiac hemodynamics in type 2 diabetes. Eur J Endocrinol 146:687 694 Lee HY, Noh HJ, Gang JG, Xu ZG, Jeong HJ, Kang SW, Choi KH, Han DS 2002 Inducible nitric oxide synthase (iNOS) expression is increased in lipopolysaccharide (LPS)-stimulated diabetic rat glomeruli: effect of ACE inhibitor and angiotensin II receptor blocker. Yonsei Med J 43:183192 Sugimoto H, Shikata K, Wada J, Horiuchi S, Makino H 1999 Advanced glycation end products-cytokine-nitric oxide sequence pathway in the development of diabetic nephropathy: aminoguanidine ameliorates the overexpression of tumour necrosis factor-

223.

224.

225.

226. 227.

228. 229.

230.

231. 232. 233.

234.

235. 236. 237. 238. 239. 240. 241.

242. 243.

nitric oxide by high glucose in cultured murine mesangial cells. Am J Physiol Renal Physiol 281:F179 F188 Cosentino F, Hishikawa K, Katusic ZS, Luscher TF 1997 High glucose increases nitric oxide synthase expression and superoxide anion generation in human aortic endothelial cells. Circulation 96:2528 Noh H, Ha H, Yu MR, Kang SW, Choi KH, Han DS, Lee HY 2002 High glucose increases inducible NO production in cultured rat mesangial cells. Possible role in fibronectin production. Nephron 90:78 85 Trachtman H, Koss I, Bogart M, Abramowitz J, Futterweit S, Franki N, Singhal PC 1998 High glucose enhances growth factorstimulated nitric oxide production by cultured rat mesangial cells. Res Commun Mol Pathol Pharmacol 100:213225 Studer RK, Craven PA, DeRubertis FR 1994 Thromboxane stimulation of mesangial cell fibronectin synthesis is signalled by protein kinase C and modulated by cGMP. Kidney Int 46:1074 1082 Keynan S, Hirshberg B, Levin-Iaina N, Wexler ID, Dahan R, Reinhartz E, Ovadia H, Wollman Y, Chernihovskey T, Iaina A, Raz I 2000 Renal nitric oxide production during the early phase of experimental diabetes mellitus. Kidney Int 58:740 747 Veelken R, Hilgers KF, Hartner A, Haas A, Bohmer KP, Sterzel RB 2000 Nitric oxide synthase isoforms and glomerular hyperfiltration in early diabetic nephropathy. J Am Soc Nephrol 11:7179 Sugimoto H, Shikata K, Matsuda M, Kushiro M, Hayashi Y, Hiragushi K, Wada J, Makino H 1998 Increased expression of endothelial cell nitric oxide synthase (ecNOS) in afferent and glomerular endothelial cells is involved in glomerular hyperfiltration of diabetic nephropathy. Diabetologia 41:1426 1434 Ishii N, Patel KP, Lane PH, Taylor T, Bian K, Murad F, Pollock JS, Carmines PK 2001 Nitric oxide synthesis and oxidative stress in the renal cortex of rats with diabetes mellitus. J Am Soc Nephrol 12:1630 1639 Schwartz D, Schwartz IF, Blantz RC 2001 An analysis of renal nitric oxide contribution to hyperfiltration in diabetic rats. J Lab Clin Med 137:107114 Onozato ML, Tojo A, Goto A, Fujita T, Wilcox CS 2002 Oxidative stress and nitric oxide synthase in rat diabetic nephropathy: effects of ACEI and ARB. Kidney Int 61:186 194 Shin SJ, Lai FJ, Wen JD, Hsiao PJ, Hsieh MC, Tzeng TF, Chen HC, Guh JY, Tsai JH 2000 Neuronal and endothelial nitric oxide synthase expression in outer medulla of streptozotocin-induced diabetic rat kidney. Diabetologia 43:649 659 De Vriese AS, Stoenoiu MS, Elger M, Devuyst O, Vanholder R, Kriz W, Lameire NH 2001 Diabetes-induced microvascular dysfunction in the hydronephrotic kidney: role of nitric oxide. Kidney Int 60:202210 Bank N, Aynedjian HS 1993 Role of EDRF (nitric oxide) in diabetic renal hyperfiltration. Kidney Int 43:1306 1312 Komers R, Allen TJ, Cooper ME 1994 Role of endothelium-derived nitric oxide in the pathogenesis of the renal hemodynamic changes of experimental diabetes. Diabetes 43:1190 1197 Reyes AA, Karl IE, Kissane J, Klahr S 1993 L-arginine administration prevents glomerular hyperfiltration and decreases proteinuria in diabetic rats. J Am Soc Nephrol 4:1039 1045 Tolins JP, Shultz PJ, Raij L, Brown DM, Mauer SM 1993 Abnormal renal hemodynamic response to reduced renal perfusion pressure in diabetic rats: role of NO. Am J Physiol 265:F886 F895 Wang YX, Brooks DP, Edwards RM 1993 Attenuated glomerular cGMP production and renal vasodilation in streptozotocininduced diabetic rats. Am J Physiol 264:R952R956 Pflueger AC, Larson TS, Hagl S, Knox FG 1999 Role of nitric oxide in intrarenal hemodynamics in experimental diabetes mellitus in rats. Am J Physiol 277:R725R733 Craven PA, Studer RK, DeRubertis FR 1994 Impaired nitric oxidedependent cyclic guanosine monophosphate generation in glomeruli from diabetic rats. Evidence for protein kinase C-mediated suppression of the cholinergic response. J Clin Invest 93:311320 De Vriese AS, Verbeuren TJ, Van de Voorde J, Lameire NH, Vanhoutte PM 2000 Endothelial dysfunction in diabetes. Br J Pharmacol 130:963974 De Vriese AS, Van de Voorde J, Blom HJ, Vanhoutte PM, Verbeke M, Lameire NH 2000 The impaired renal vasodilator response

244.

245.

246.

247.

248.

249. 250. 251.

252.

253.

254. 255. 256.

257. 258.

259.

260.

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy and inducible nitric oxide synthase in diabetic rat glomeruli. Diabetologia 42:878 886 De Vriese AS, Tilton RG, Elger M, Stephan CC, Kriz W, Lameire NH 2001 Antibodies against vascular endothelial growth factor improve early renal dysfunction in experimental diabetes. J Am Soc Nephrol 12:9931000 Webb BLJ, Hirst SJ, Giembycz MA 2000 Protein kinase C isoenzymes: a review of their structure, regulation and role in regulating airways smooth muscle tone and mitogenesis. Br J Pharmacol 130: 14331452 Way KJ, Chou E, King GL 2000 Identification of PKC-isoformspecific biological actions using pharmacological approaches. Trends Pharmacol Sci 21:181187 Idris I, Gray S, Donnelly R 2001 Protein kinase C activation: isozyme-specific effects on metabolism and cardiovascular complications in diabetes. Diabetologia 44:659 673 Mochly-Rosen D, Gordon AS 1998 Anchoring proteins for protein kinase C: a means for isozyme selectivity. FASEB J 12:35 42 Koya D, King GL 1998 Protein kinase C in diabetic renal involvement, the perspective of its inhibition. In: Mogensen CE, ed. The kidney and hypertension in diabetes mellitus. Boston, Dordrecht, London: Kluwer Academic Publishers; 263268 Johannes FJ, Prestle J, Eis S, Oberhagemann P, Pfizenmaier K 1994 PKCu is a novel, atypical member of the protein kinase C family. J Biol Chem 269:6140 6148 Amiri F, Garcia R 2000 Renal angiotensin II receptors and protein kinase C in diabetic rats: effects of insulin and ACE inhibition. Am J Physiol Renal Physiol 278:F603F612 Xia P, Aiello LP, Ishii H, Jiang ZY, Park DJ, Robinson GS, Takagi H, Newsome WP, Jirousek MR, King GL 1996 Characterization of vascular endothelial growth factors effect on the activation of protein kinase C, its isoforms, and endothelial cell growth. J Clin Invest 98:2018 2026 Promeneur D, Frokir J, Nielsen S 2002 Immunolocalization of the PKC isoforms , , and in rat kidney. J Am Soc Nephrol 13:290A (Abstract) Saxena R, Saksa BA, Hawkins KS, Ganz MB 1994 Protein kinase C I and II are differentially expressed in the developing glomerulus. FASEB J 8:646 653 Koya D, Jirousek MR, Lin YW, Ishii H, Kuboki K, King GL 1997 Characterization of protein kinase C isoform activation on the gene expression of transforming growth factor-, extracellular matrix components, and prostanoids in the glomeruli of diabetic rats. J Clin Invest 100:115126 Isshiki K, Haneda M, Koya D, Maeda S, Sugimoto T, Kikkawa R 2000 Thiazolidinedione compounds ameliorate glomerular dysfunction independent of their insulin-sensitizing action in diabetic rats. Diabetes 49:10221032 Craven PA, DeRubertis FR 1989 Protein kinase C is activated in glomeruli from streptozotocin diabetic rats. Possible mediation by glucose. J Clin Invest 83:16671675 Keogh RJ, Dunlop ME, Larkins RG 1997 Effect of inhibition of aldose reductase on glucose flux, diacylglycerol formation, protein kinase C, and phospholipase A2 activation. Metabolism 46:41 47 Park CW, Kim JH, Lee JW, Kim YS, Ahn HJ, Shin YS, Kim SY, Choi EJ, Chang YS, Bang BK 2000 High glucose-induced intercellular adhesion molecule-1 (ICAM-1) expression through an osmotic effect in rat mesangial cells is PKC-NF- B-dependent. Diabetologia 43:1544 1553 Awazu M, Ishikura K, Hida M, Hoshiya M 1999 Mechanisms of mitogen-activated protein kinase activation in experimental diabetes. J Am Soc Nephrol 10:738 745 Ganz MB, Saksa B, Saxena R, Hawkins K, Sedor JR 1996 PDGF and IL-1 induce and activate specific protein kinase C isoforms in mesangial cells. Am J Physiol 271:F108 F113 Williams B, Gallacher B, Patel H, Orme C 1997 Glucose-induced protein kinase C activation regulates vascular permeability factor mRNA expression and peptide production by human vascular smooth muscle cells in vitro. Diabetes 46:14971503 Koya D, Lee IK, Ishii H, Kanoh H, King GL 1997 Prevention of glomerular dysfunction in diabetic rats by treatment with d-tocopherol. J Am Soc Nephrol 8:426 435 Ishii H, Jirousek MR, Koya D, Takagi C, Xia P, Clermont A,

Endocrine Reviews, December 2004, 25(6):9711010 1005 Bursell SE, Kern TS, Ballas LM, Heath WF, Stramm LE, Feener EP, King GL 1996 Amelioration of vascular dysfunctions in diabetic rats by an oral PKC inhibitor. Science 272:728 731 Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, King GL, Kikkawa R 2000 Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J 14:439 447 Araki S, Ng DP, Krolewski B, Wyrwicz L, Rogus JJ, Canani L, Makita Y, Haneda M, Warram JH, Krolewski AS 2003 Identification of a common risk haplotype for diabetic nephropathy at the protein kinase C-1 (PRKCB1) gene locus. J Am Soc Nephrol 14: 20152024 Ceolotto G, Gallo A, Miola M, Sartori M, Trevisan R, Del Prato S, Semplicini A, Avogaro A 1999 Protein kinase C activity is acutely regulated by plasma glucose concentration in human monocytes in vivo. Diabetes 48:1316 1322 Machiguchi T, Kimura H, Yonemoto S, Kimura N, Li X, Suzuki S, Yoshida H 2002 Glomerular expression of protein kinase C (PKC) and PCNA in human diabetic nephropathy. J Am Soc Nephrol 13:647A (Abstract) Haller H, Maasch C, Dragun D, Wellner M, Janta-Lipinski M, Luft FC 1998 Antisense oligodesoxynucleotide strategies in renal and cardiovascular disease. Kidney Int 53:1550 1558 Kelly DJ, Zhang Y, Hepper C, Gow RM, Jaworski K, Kemp BE, Wilkinson-Berka JL, Gilbert RE 2003 Protein kinase C inhibition attenuates the progression of experimental diabetic nephropathy in the presence of continued hypertension. Diabetes 52:512518 Flyvbjerg A, Hill C, Nielsen B, Logan A 1999 Effect of protein kinase C inhibition on renal morphology, urinary albumin excretion and renal transforming growth factor in experimental diabetes in rats. J Am Soc Nephrol 10:A3445 (Abstract) Tuttle KR, Anderson PW 2003 A novel potential therapy for diabetic nephropathy and vascular complications: protein kinase C inhibition. Am J Kidney Dis 42:456 465 Tran K, Proulx PR, Chan AC 1994 Vitamin E suppresses diacylglycerol (DAG) level in thrombin-stimulated endothelial cells through an increase of DAG kinase activity. Biochim Biophys Acta 1212:193202 Lee IK, Koya D, Ishi H, Kanoh H, King GL 1999 d--tocopherol prevents the hyperglycemia induced activation of diacylglycerol (DAG)-protein kinase C (PKC) pathway in vascular smooth muscle cell by an increase of DAG kinase activity. Diabetes Res Clin Pract 45:183190 Koya D, Haneda M, Kikkawa R, King GL 1998 d--Tocopherol treatment prevents glomerular dysfunctions in diabetic rats through inhibition of protein kinase C-diacylglycerol pathway. Biofactors 7:69 76 Yoshimoto T, Naruse M, Nishikawa M, Naruse K, Tanabe A, Seki T, Imaki T, Demura R, Aikawa E, Demura H 1997 Antihypertensive and vasculo- and renoprotective effects of pioglitazone in genetically obese diabetic rats. Am J Physiol 272:E989 E996 Routh RE, Johnson JH, McCarthy KJ 2002 Troglitazone suppresses the secretion of type I collagen by mesangial cells in vitro. Kidney Int 61:13651376 Piek E, Heldin CH, Ten Dijke P 1999 Specificity, diversity, and regulation in TGF- superfamily signaling. FASEB J 13:21052124 Wrana JL, Attisano L, Wieser R, Ventura F, Massague J 1994 Mechanism of activation of the TGF- receptor. Nature 370:341347 Kloos DU, Choi C, Wingender E 2002 The TGF-Smad network: introducing bioinformatic tools. Trends Genet 18:96 103 Riser BL, Denichilo M, Cortes P, Baker C, Grondin JM, Yee J, Narins RG 2000 Regulation of connective tissue growth factor activity in cultured rat mesangial cells and its expression in experimental diabetic glomerulosclerosis. J Am Soc Nephrol 11:2538 Hill C, Flyvbjerg A, Grnbk H, Petrik J, Hill DJ, Thomas CR, Sheppard MC, Logan A 2000 The renal expression of transforming growth factor- isoforms and their receptors in acute and chronic experimental diabetes in rats. Endocrinology 141:1196 1208 Nakamura T, Fukui M, Ebihara I, Osada S, Nagaoka I, Tomino Y, Koide H 1993 mRNA expression of growth factors in glomeruli from diabetic rats. Diabetes 42:450 456 Gilbert RE, Cox A, Wu LL, Allen TJ, Hulthen UL, Jerums G,

261.

282.

262.

283.

263. 264. 265. 266.

284.

285.

286. 287.

267. 268. 269.

288.

289. 290.

270. 271. 272.

291.

273.

292.

274. 275. 276.

293.

294. 295. 296. 297. 298.

277. 278. 279.

299.

280. 281.

300. 301.

Downloaded from edrv.endojournals.org on April 19, 2006

1006

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy 320. Kelly FJ, Anderson S, Thompson MM, Oyama TT, Kennefick TM, Corless CL, Roman RJ, Kurtzberg L, Pratt BM, Ledbetter SR 1999 Acute and chronic renal effects of recombinant human TGF-2 in the rat. J Am Soc Nephrol 10:1264 1273 321. Azar ST, Salti I, Zantout MS, Major S 2000 Alterations in plasma transforming growth factor in normoalbuminuric type 1 and type 2 diabetic patients. J Clin Endocrinol Metab 85:4680 4682 322. Korpinen E, Teppo AM, Hukkanen L, kerblom HK, Gro nhagen-Riska C, Vaarala O 2000 Urinary transforming growth factor-1 and 1-microglobulin in children and adolescents with type 1 diabetes. Diabetes Care 23:664 668 323. Ellis D, Forrest KY, Erbey J, Orchard TJ 1998 Urinary measurement of transforming growth factor- and type IV collagen as new markers of renal injury: application in diabetic nephropathy. Clin Chem 44:950 956 324. Yamamoto T, Noble NA, Cohen AH, Nast CC, Hishida A, Gold LI, Border WA 1996 Expression of transforming growth factor- isoforms in human glomerular diseases. Kidney Int 49:461 469 325. Yamamoto T, Nakamura T, Noble NA, Ruoslahti E, Border WA 1993 Expression of transforming growth factor- is elevated in human and experimental diabetic nephropathy. Proc Natl Acad Sci USA 90:1814 1818 326. Sharma K, Ziyadeh FN, Alzahabi B, McGowan TA, Kapoor S, Kurnik BR, Kurnik PB, Weisberg LS 1997 Increased renal production of transforming growth factor-1 in patients with type II diabetes. Diabetes 46:854 859 327. Hellmich B, Schellner M, Schatz H, Pfeiffer A 2000 Activation of transforming growth factor-1 in diabetic kidney disease. Metabolism 49:353359 328. Sato H, Iwano M, Akai Y, Kurioka H, Kubo A, Yamaguchi T, Hirata E, Kanauchi M, Dohi K 1998 Increased excretion of urinary transforming growth factor 1 in patients with diabetic nephropathy. Am J Nephrol 18:490 494 329. Mogyorosi A, Kapoor A, Isono M, Kapoor S, Sharma K, Ziyadeh FN 2000 Utility of serum and urinary transforming growth factor- levels as markers of diabetic nephropathy. Nephron 86:234 235 330. Iwano M, Kubo A, Nishino T, Sato H, Nishioka H, Akai Y, Kurioka H, Fujii Y, Kanauchi M, Shiiki H, Dohi K 1996 Quantification of glomerular TGF- 1 mRNA in patients with diabetes mellitus. Kidney Int 49:1120 1126 331. Ito Y, Aten J, Bende RJ, Oemar BS, Rabelink TJ, Weening JJ, Goldschmeding R 1998 Expression of connective tissue growth factor in human renal fibrosis. Kidney Int 53:853 861 332. Sharma K, Jin Y, Guo J, Ziyadeh FN 1996 Neutralization of TGF- by anti-TGF- antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in STZ-induced diabetic mice. Diabetes 45:522530 333. Hill C, Flyvbjerg A, Rasch R, Bak M, Logan A 2001 Transforming growth factor-2 antibody attenuates fibrosis in the experimental diabetic rat kidney. J Endocrinol 170:647 651 334. Ziyadeh FN, Hoffman BB, Han DC, Iglesias-De La Cruz MC, Hong SW, Isono M, Chen S, McGowan TA, Sharma K 2000 Long-term prevention of renal insufficiency, excess matrix gene expression, and glomerular mesangial matrix expansion by treatment with monoclonal antitransforming growth factor- antibody in db/db diabetic mice. Proc Natl Acad Sci USA 97:8015 8020 335. Han DC, Hoffman BB, Hong SW, Guo J, Ziyadeh FN 2000 Therapy with antisense TGF-1 oligodeoxynucleotides reduces kidney weight and matrix mRNAs in diabetic mice. Am J Physiol Renal Physiol 278:F628 F634 336. Hill C, Logan A, Smith C, Grnbk H, Flyvbjerg A 2001 Angiotensin converting enzyme inhibitor suppresses glomerular transforming growth factor- receptor expression in experimental diabetes in rats. Diabetologia 44:495500 337. Sharma K, Eltayeb BO, McGowan TA, Dunn SR, Alzahabi B, Rohde R, Ziyadeh FN, Lewis EJ 1999 Captopril-induced reduction of serum levels of transforming growth factor-1 correlates with long-term renoprotection in insulin-dependent diabetic patients. Am J Kidney Dis 34:818 823 338. Kim SI, Han DC, Lee HB 2000 Lovastatin inhibits transforming growth factor-1 expression in diabetic rat glomeruli and cultured rat mesangial cells. J Am Soc Nephrol 11:80 87 339. Goppelt-Struebe M, Hahn A, Iwanciw D, Rehm M, Banas B 2001

302.

303.

304. 305. 306.

307.

308.

309. 310. 311. 312. 313.

314. 315. 316.

317.

318.

319.

Cooper ME 1998 Expression of transforming growth factor-1 and type IV collagen in the renal tubulointerstitium in experimental diabetes: effects of ACE inhibition. Diabetes 47:414 422 Hong SW, Isono M, Chen S, Iglesias-De La Cruz MC, Han DC, Ziyadeh FN 2001 Increased glomerular and tubular expression of transforming growth factor-1, its type II receptor, and activation of the Smad signaling pathway in the db/db mouse. Am J Pathol 158:16531663 Fujigaki Y, Watanabe T, Ikegaya N, Yonemura K, Sun DF, Hishida A, Yamamoto T, Kojima K, Nagase M 2000 Immunoelectron microscopic study on type I, II and III TGF- receptors on visceral glomerular epithelial cells in relation to glomerular basement membrane alterations in proteinuric rats. Nephrol Dial Transplant 15:191199 Wang S, Denichilo M, Brubaker C, Hirschberg R 2001 Connective tissue growth factor in tubulointerstitial injury of diabetic nephropathy. Kidney Int 60:96 105 Tack I, Elliot SJ, Potier M, Rivera A, Striker GE, Striker LJ 2002 Autocrine activation of the IGF-I signaling pathway in mesangial cells isolated from diabetic NOD mice. Diabetes 51:182188 Kagami S, Border WA, Miller DE, Noble NA 1994 Angiotensin II stimulates extracellular matrix protein synthesis through induction of transforming growth factor- expression in rat glomerular mesangial cells. J Clin Invest 93:24312437 Ziyadeh FN, Sharma K, Ericksen M, Wolf G 1994 Stimulation of collagen gene expression and protein synthesis in murine mesangial cells by high glucose is mediated by autocrine activation of transforming growth factor-. J Clin Invest 93:536 542 Davies M, Thomas GJ, Martin J, Lovett DH 1988 The purification and characterization of a glomerular-basement-membrane-degrading neutral proteinase from rat mesangial cells. Biochem J 251: 419 425 Marti HP, Lee L, Kashgarian M, Lovett DH 1994 Transforming growth factor-1 stimulates glomerular mesangial cell synthesis of the 72-kd type IV collagenase. Am J Pathol 144:8294 Nakamura T, Miller D, Ruoslahti E, Border WA 1992 Production of extracellular matrix by glomerular epithelial cells is regulated by transforming growth factor-1. Kidney Int 41:12131221 Wang SN, Hirschberg R 2000 Growth factor ultrafiltration in experimental diabetic nephropathy contributes to interstitial fibrosis. Am J Physiol Renal Physiol 278:F554 F560 Wang SN, Lapage J, Hirschberg R 2000 Role of glomerular ultrafiltration of growth factors in progressive interstitial fibrosis in diabetic nephropathy. Kidney Int 57:10021014 Blom IE, van Dijk AJ, Wieten L, Duran K, Ito Y, Kleij L, Denichilo M, Rabelink TJ, Weening JJ, Aten J, Goldschmeding R 2001 In vitro evidence for differential involvement of CTGF, TGF, and PDGF-BB in mesangial response to injury. Nephrol Dial Transplant 16:1139 1148 Shankland SJ, Scholey JW, Ly H, Thai K 1994 Expression of transforming growth factor-1 during diabetic renal hypertrophy. Kidney Int 46:430 442 Sharma K, Ziyadeh FN 1994 Renal hypertrophy is associated with upregulation of TGF- 1 gene expression in diabetic BB rat and NOD mouse. Am J Physiol 267:F1094 F1101 Yang CW, Hattori M, Vlassara H, He CJ, Carome MA, Yamato E, Elliot S, Striker GE, Striker LJ 1995 Overexpression of transforming growth factor- 1 mRNA is associated with up-regulation of glomerular tenascin and laminin gene expression in nonobese diabetic mice. J Am Soc Nephrol 5:1610 1617 Montero A, Munger KA, Khan RZ, Valdivielso JM, Morrow JD, Guasch A, Ziyadeh FN, Badr KF 2000 F(2)-isoprostanes mediate high glucose-induced TGF- synthesis and glomerular proteinuria in experimental type I diabetes. Kidney Int 58:19631972 Aoyama I, Shimokata K, Niwa T 2000 Oral adsorbent AST-120 ameliorates interstitial fibrosis and transforming growth factor(1) expression in spontaneously diabetic (OLETF) rats. Am J Nephrol 20:232241 Tsuchida K, Makita Z, Yamagishi S, Atsumi T, Miyoshi H, Obara S, Ishida M, Ishikawa S, Yasumura K, Koike T 1999 Suppression of transforming growth factor and vascular endothelial growth factor in diabetic nephropathy in rats by a novel advanced glycation end product inhibitor, OPB-9195. Diabetologia 42:579 588

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy Regulation of connective tissue growth factor (ccn2; ctgf) gene expression in human mesangial cells: modulation by HMG CoA reductase inhibitors (statins). Mol Pathol 54:176 179 Studer RK, Craven PA, DeRubertis FR 1997 Antioxidant inhibition of protein kinase C-signaled increases in transforming growth factor- in mesangial cells. Metabolism 46:918 925 Craven PA, DeRubertis FR, Kagan VE, Melhem M, Studer RK 1997 Effects of supplementation with vitamin C or E on albuminuria, glomerular TGF-, and glomerular size in diabetes. J Am Soc Nephrol 8:14051414 Aoyama I, Shimokata K, Niwa T 2002 Combination therapy with benazepril and oral adsorbent ameliorates progressive renal fibrosis in uremic rats. Nephron 90:297312 Ballesteros M, Leung KC, Ross RJM, Iismaa TP, Ho KKY 2000 Distribution and abundance of messenger ribonucleic acid for growth hormone receptor isoforms in human tissues. J Clin Endocrinol Metab 85:28652871 Zhu T, Goh EL, Graichen R, Ling L, Lobie PE 2001 Signal transduction via the growth hormone receptor. Cell Signal 13:599 616 Baumann G 2001 Growth hormone binding protein 2001. J Pediatr Endocrinol Metab 14:355375 Hwa V, Oh Y, Rosenfeld RG 1999 The insulin-like growth factorbinding protein (IGFBP) superfamily. Endocr Rev 20:761787 LeRoith D 2000 Insulin-like growth factor I receptor signaling-overlapping or redundant pathways? Endocrinology 141:12871288 Werner H, Shen-Orr Z, Stannard B, Burguera B, Roberts Jr CT, LeRoith D 1990 Experimental diabetes increases insulin like growth factor I and II receptor concentration and gene expression in kidney. Diabetes 39:1490 1497 Flyvbjerg A, Nielsen S, Sheikh MI, Jacobsen C, rskov H, Christensen EI 1993 Luminal and basolateral uptake and receptor binding of IGF-I in rabbit renal proximal tubules. Am J Physiol 265: F624 F633 Feld S, Hirschberg R 1996 Growth hormone, the insulin-like growth factor system, and the kidney. Endocr Rev 17:423 480 Chin E, Zhou J, Bondy CA 1992 Renal growth hormone receptor gene expression: relationship to renal insulin-like growth factor system. Endocrinology 131:30613066 Sayed-Ahmed N, Muchaneta-Kubara EC, Besbas N, Shortland J, Cope GH, El Nahas AM 1993 Insulin-like growth factor-I and experimental diabetic kidney disease. Exp Nephrol 1:364 371 Chin E, Zhou J, Bondy C 1992 Anatomical relationships in the patterns of insulin-like growth factor (IGF)-I, IGF binding protein-1, and IGF-I receptor gene expression in the rat kidney. Endocrinology 130:32373245 Conti FG, Striker LJ, Elliot SJ, Andreani D, Striker GE 1988 Synthesis and release of insulin like growth factor I by mesangial cells in culture. Am J Physiol 255:F1214 F1219 Conti FG, Striker LJ, Lesniak MA, MacKay K, Roth J, Striker GE 1988 Studies on binding and mitogenic effect of insulin and insulinlike growth factor I in glomerular mesangial cells. Endocrinology 122:2788 2795 Conti FG, Elliot SJ, Striker LJ, Striker GE 1989 Binding of insulinlike growth factor-I by glomerular endothelial and epithelial cells: further evidence for IGF-I action in the renal glomerulus. Biochem Biophys Res Commun 163:952958 Doi T, Striker LJ, Elliot SJ, Conti FG, Striker GE 1989 Insulin like growth factor-1 is a progression factor for human mesangial cells. Am J Pathol 134:395 404 Schreiber BD, Hughes ML, Groggel GC 1995 Insulin-like growth factor-1 stimulates production of mesangial cell matrix components. Clin Nephrol 43:368 374 Moran A, Brown DM, Kim Y, Klein DJ 1991 Effects of IGF-I and glucose on protein and proteoglycan synthesis by human fetal mesangial cells in culture. Diabetes 40:1346 1354 Oemar BS, Foellmer HG, Hodgdon-Anandant L, Rosenzweig SA 1991 Regulation of insulin-like growth factor I receptors in diabetic mesangial cells. J Biol Chem 266:2369 2373 Flyvbjerg A 2000 Putative pathophysiological role of growth factors and cytokines in experimental diabetic kidney disease. Diabetologia 43:12051223 Doi T, Striker LJ, Quaife C, Conti FG, Palmiter R, Behringer R, Brinster R, Striker GE 1988 Progressive glomerulosclerosis devel-

Endocrine Reviews, December 2004, 25(6):9711010 1007 ops in transgenic mice chronically expressing growth hormone and growth hormone releasing factor but not in those expressing insulin like growth factor-1. Am J Pathol 131:398 403 Doi T, Striker LJ, Gibson CC, Agodoa LY, Brinster RL, Striker GE 1990 Glomerular lesions in mice transgenic for growth hormone and insulin like growth factor-I. I. Relationship between increased glomerular size and mesangial sclerosis. Am J Pathol 137:541552 Tannenbaum GS 1981 Growth hormone secretory dynamics in streptozotocin diabetes: evidence of a role for endogenous circulating somatostatin. Endocrinology 108:76 82 Mller N, rskov H 2000 Growth hormone, IGF-I and diabetic angiopathy revisited. Clin Endocrinol (Oxf) 52:1112 Bereket A, Lang CH, Wilson TA 1999 Alterations in the growth hormone-insulin-like growth factor axis in insulin dependent diabetes mellitus. Horm Metab Res 31:172181 Flyvbjerg A, Kessler U, Dorka B, Funk B, rskov H, Kiess W 1992 Transient increase in renal insulin-like growth factor binding proteins during initial kidney hypertrophy in experimental diabetes in rats. Diabetologia 35:589 593 Flyvbjerg A 1997 Role of growth hormone, insulin-like growth factors (IGFs) and IGF-binding proteins in the renal complications of diabetes. Kidney Int Suppl 60:S12S19 Flyvbjerg A, Bennett WF, Rasch R, Kopchick JJ, Scarlett JA 1999 Inhibitory effect of a growth hormone receptor antagonist (G120KPEG) on renal enlargement, glomerular hypertrophy, and urinary albumin excretion in experimental diabetes in mice. Diabetes 48: 377382 Massa G, Verhaeghe J, Vanderschueren-Lodeweyckx M, Bouillon R 1993 Normalization of decreased plasma concentrations of growth hormone- binding protein by insulin treatment in spontaneously diabetic BB rats. Horm Metab Res 25:325326 Landau D, Domene H, Flyvbjerg A, Grnbk H, Roberts Jr CT, Argov S, LeRoith D 1998 Differential expression of renal growth hormone receptor and its binding protein in experimental diabetes mellitus. Growth Horm IGF Res 8:39 45 Segev Y, Landau D, Marbach M, Shehadeh N, Flyvbjerg A, Phillip M 1997 Renal hypertrophy in hyperglycemic non-obese diabetic mice is associated with persistent renal accumulation of insulin-like growth factor I. J Am Soc Nephrol 8:436 444 Park IS, Kiyomoto H, Alvarez F, Xu YC, Abboud HE, Abboud SL 1998 Preferential expression of insulin-like growth factor binding proteins-1, -3, and -5 during early diabetic renal hypertrophy in rats. Am J Kidney Dis 32:1000 1010 Flyvbjerg A, Kessler U, Kiess W 1994 Increased kidney and liver insulin-like growth factor II/mannose-6- phosphate receptor concentration in experimental diabetes in rats. Growth Regul 4: 188 193 Bach LA, Jerums G 1990 Effect of puberty on initial kidney growth and rise in kidney IGF-I in diabetic rats. Diabetes 39:557562 Flyvbjerg A, Frystyk J, sterby R, rskov H 1992 Kidney IGF-I and renal hypertrophy in GH-deficient diabetic dwarf rats. Am J Physiol 262:E956 E962 Grnbk H, Volmers P, Bjorn SF, sterby R, rskov H, Flyvbjerg A 1997 Effect of GH/IGF-I deficiency on long-term renal changes and urinary albumin excretion in diabetic dwarf rats. Am J Physiol 272:E918 E924 Verrotti A, Cieri F, Petitti MT, Morgese G, Chiarelli F 1999 Growth hormone and IGF-I in diabetic children with and without microalbuminuria. Diabetes Nutr Metab 12:271276 Sen A, Buyukgebiz A 1997 Albumin excretion rate, serum insulinlike growth factor-I and glomerular filtration rate in type I diabetes mellitus at puberty. J Pediatr Endocrinol Metab 10:209 215 Cummings EA, Sochett EB, Dekker MG, Lawson ML, Daneman D 1998 Contribution of growth hormone and IGF-I to early diabetic nephropathy in type 1 diabetes. Diabetes 47:13411346 Bacci S, De Cosmo S, Garruba M, Placentino G, Liuzzi A, Barbano F, Di Giorgio A, Trischitta V, Viberti GC 2000 Role of insulin-like growth factor (IGF)-1 in the modulation of renal haemodynamics in type I diabetic patients. Diabetologia 43:922926 Hoogenberg K, ter Wee PM, Lieverse AG, Sluiter WJ, Dullaart RP 1994 Insulin-like growth factor I and altered renal hemodynamics in growth hormone deficiency, acromegaly, and type I diabetes mellitus. Transplant Proc 26:505507

340. 341.

363.

364. 365. 366. 367.

342. 343.

344. 345. 346. 347. 348.

368. 369.

370.

349.

371.

350. 351. 352. 353.

372.

373.

374.

354. 355.

375. 376. 377.

356.

357. 358. 359. 360. 361. 362.

378. 379. 380. 381.

382.

Downloaded from edrv.endojournals.org on April 19, 2006

1008

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy uli, and the identification of VEGF148 mRNA, a novel truncated splice variant. Clin Sci (Lond) 97:303312 Hood JD, Meininger CJ, Ziche M, Granger HJ 1998 VEGF upregulates ecNOS message, protein, and NO production in human endothelial cells. Am J Physiol 274:H1054 H1058 Cooper ME, Vranes D, Youssef S, Stacker SA, Cox AJ, Rizkalla B, Casley DJ, Bach LA, Kelly DJ, Gilbert RE 1999 Increased renal expression of vascular endothelial growth factor (VEGF) and its receptor VEGFR-2 in experimental diabetes. Diabetes 48:2229 2239 Simon M, Ro ckl W, Hornig C, Gro ne EF, Theis H, Weich HA, Fuchs E, Yayon A, Gro ne HJ 1998 Receptors of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) in fetal and adult human kidney: localization and [125I]VEGF binding sites. J Am Soc Nephrol 9:10321044 Simon M, Gro ne HJ, Johren O, Kullmer J, Plate KH, Risau W, Fuchs E 1995 Expression of vascular endothelial growth factor and its receptors in human renal ontogenesis and in adult kidney. Am J Physiol 268:F240 F250 Cha DR, Kim NH, Yoon JW, Jo SK, Cho WY, Kim HK, Won NH 2000 Role of vascular endothelial growth factor in diabetic nephropathy. Kidney Int Suppl 77:S104 S112 Iijima K, Yoshikawa N, Connolly DT, Nakamura H 1993 Human mesangial cells and peripheral blood mononuclear cells produce vascular permeability factor. Kidney Int 44:959 966 Kretzler M, Schro ppel B, Merkle M, Huber S, Mundel P, Horster M, Schlo ndorff D 1998 Detection of multiple vascular endothelial growth factor splice isoforms in single glomerular podocytes. Kidney Int Suppl 67:S159 S161 Amemiya T, Sasamura H, Mifune M, Kitamura Y, Hirahashi J, Hayashi M, Saruta T 1999 Vascular endothelial growth factor activates MAP kinase and enhances collagen synthesis in human mesangial cells. Kidney Int 56:20552063 Thomas S, Vanuystel J, Gruden G, Rodriguez V, Burt D, Gnudi L, Hartley B, Viberti G 2000 Vascular endothelial growth factor receptors in human mesangium in vitro and in glomerular disease. J Am Soc Nephrol 11:1236 1243 Trachtman H, Futterweit S, Franki N, Singhal PC 1998 Effect of vascular endothelial growth factor on nitric oxide production by cultured rat mesangial cells. Biochem Biophys Res Commun 245: 443 446 Kanellis J, Fraser S, Katerelos M, Power DA 2000 Vascular endothelial growth factor is a survival factor for renal tubular epithelial cells. Am J Physiol Renal Physiol 278:F905F915 Harper SJ, Xing CY, Whittle C, Parry R, Gillatt D, Peat D, Mathieson PW 2001 Expression of neuropilin-1 by human glomerular epithelial cells in vitro and in vivo. Clin Sci (Lond) 101:439 446 El Awad B, Kreft B, Wolber EM, Hellwig-Burgel T, Metzen E, Fandrey J, Jelkmann W 2000 Hypoxia and interleukin-1 stimulate vascular endothelial growth factor production in human proximal tubular cells. Kidney Int 58:4350 Gruden G, Thomas S, Burt D, Zhou W, Chusney G, Gnudi L, Viberti G 1999 Interaction of angiotensin II and mechanical stretch on vascular endothelial growth factor production by human mesangial cells. J Am Soc Nephrol 10:730 737 Pupilli C, Lasagni L, Romagnani P, Bellini F, Mannelli M, Misciglia N, Mavilia C, Vellei U, Villari D, Serio M 1999 Angiotensin II stimulates the synthesis and secretion of vascular permeability factor/vascular endothelial growth factor in human mesangial cells. J Am Soc Nephrol 10:245255 Kim NH, Jung HH, Cha DR, Choi DS 2000 Expression of vascular endothelial growth factor in response to high glucose in rat mesangial cells. J Endocrinol 165:617 624 Yamagishi S, Inagaki Y, Okamoto T, Amano S, Koga K, Takeuchi M, Makita Z 2002 Advanced glycation end product-induced apoptosis and overexpression of vascular endothelial growth factor and monocyte chemoattractant protein-1 in human-cultured mesangial cells. J Biol Chem 277:20309 20315 Mandriota SJ, Menoud PA, Pepper MS 1996 Transforming growth factor 1 down-regulates vascular endothelial growth factor receptor 2/flk-1 expression in vascular endothelial cells. J Biol Chem 271:11500 11505 Marumo T, Schini-Kerth VB, Busse R 1999 Vascular endothelial growth factor activates nuclear factor-B and induces monocyte

383. Jehle PM, Jehle DR, Mohan S, Bohm BO 1998 Serum levels of insulin-like growth factor system components and relationship to bone metabolism in type 1 and type 2 diabetes mellitus patients. J Endocrinol 159:297306 384. Shinada M, Akdeniz A, Panagiotopoulos S, Jerums G, Bach LA 2000 Proteolysis of insulin-like growth factor-binding protein-3 is increased in urine from patients with diabetic nephropathy. J Clin Endocrinol Metab 85:11631169 385. Flyvbjerg A, Frystyk J, Thorlacius-Ussing O, rskov H 1989 Somatostatin analogue administration prevents increase in kidney somatomedin C and initial renal growth in diabetic and uninephrectomized rats. Diabetologia 32:261265 386. Grnbk H, Nielsen B, Frystyk J, rskov H, Flyvbjerg A 1995 Effect of octreotide on experimental diabetic renal and glomerular growth: importance of early intervention. J Endocrinol 147:95102 387. Flyvbjerg A, Marshall SM, Frystyk J, Hansen KW, Harris AG, rskov H 1992 Octreotide administration in diabetic rats: effects on renal hypertrophy and urinary albumin excretion. Kidney Int 41: 805 812 388. Grnbk H, Nielsen B, Schrijvers B, Vogel I, Rasch R, Flyvbjerg A 2002 Inhibitory effects of octreotide on renal and glomerular growth in early experimental diabetes in mice. J Endocrinol 172: 637 643 389. Landau D, Segev Y, Afargan M, Silbergeld A, Katchko L, Podshyvalov A, Phillip M 2001 A novel somatostatin analogue prevents early renal complications in the nonobese diabetic mouse. Kidney Int 60:505512 390. Pedersen MM, Christensen SE, Christiansen JS, Pedersen EB, Mogensen CE, rskov H 1990 Acute effects of a somatostatin analogue on kidney function in type 1 diabetic patients. Diabet Med 7:304 309 391. Serri O, Beauregard H, Brazeau P, Abribat T, Lambert J, Harris A, Vachon L 1991 Somatostatin analogue, octreotide, reduces increased glomerular filtration rate and kidney size in insulindependent diabetes. JAMA 265:888 892 392. Bellush LL, Doublier S, Holland AN, Striker LJ, Striker GE, Kopchick JJ 2000 Protection against diabetes-induced nephropathy in growth hormone receptor/binding protein gene-disrupted mice. Endocrinology 141:163168 393. Chen NY, Chen WY, Bellush L, Yang CW, Striker LJ, Striker GE, Kopchick JJ 1995 Effects of streptozotocin treatment in growth hormone (GH) and GH antagonist transgenic mice. Endocrinology 136:660 667 394. Chen NY, Chen WY, Kopchick JJ 1996 A growth hormone antagonist protects mice against streptozotocin induced glomerulosclerosis even in the presence of elevated levels of glucose and glycated hemoglobin. Endocrinology 137:51635165 395. Segev Y, Landau D, Rasch R, Flyvbjerg A, Phillip M 1999 Growth hormone receptor antagonism prevents early renal changes in nonobese diabetic mice. J Am Soc Nephrol 10:2374 2381 396. Haylor J, Hickling H, El Eter E, Moir A, Oldroyd S, Hardisty C, El Nahas AM 2000 JB3, an IGF-I receptor antagonist, inhibits early renal growth in diabetic and uninephrectomized rats. J Am Soc Nephrol 11:20272035 397. New JP, Canavan JP, Flyvbjerg A, Hamon G, Bilous RW, Marshall SM 1996 Renal enlargement and insulin-like growth factor-1 accumulation in the Wistar rat model of experimental diabetes is not prevented by angiotensin converting enzyme inhibition. Diabetologia 39:166 171 398. Bach LA, Dean R, Youssef S, Cooper ME 2000 Aminoguanidine ameliorates changes in the IGF system in experimental diabetic nephropathy. Nephrol Dial Transplant 15:347354 399. Robinson CJ, Stringer SE 2001 The splice variants of vascular endothelial growth factor (VEGF) and their receptors. J Cell Sci 114:853 865 400. Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z 1999 Vascular endothelial growth factor (VEGF) and its receptors. FASEB J 13: 9 22 401. Ferrara N, Gerber HP 2001 The role of vascular endothelial growth factor in angiogenesis. Acta Haematol 106:148 156 402. Whittle C, Gillespie K, Harrison R, Mathieson PW, Harper SJ 1999 Heterogeneous vascular endothelial growth factor (VEGF) isoform mRNA and receptor mRNA expression in human glomer-

403. 404.

405.

406.

407. 408. 409.

410.

411.

412.

413. 414. 415.

416.

417.

418. 419.

420.

421.

Downloaded from edrv.endojournals.org on April 19, 2006

Schrijvers et al. Factors Influencing Diabetic Nephropathy chemoattractant protein-1 in bovine retinal endothelial cells. Diabetes 48:11311137 Saijonmaa O, Nyman T, Kosonen R, Fyhrquist F 2001 Upregulation of angiotensin-converting enzyme by vascular endothelial growth factor. Am J Physiol Heart Circ Physiol 280:H885H891 Senthil D, Choudhury GG, McLaurin C, Kasinath BS 2003 Vascular endothelial growth factor induces protein synthesis in renal epithelial cells: a potential role in diabetic nephropathy. Kidney Int 64:468 479 Cheng HF, Wang CJ, Moeckel GW, Zhang MZ, McKanna JA, Harris RC 2002 Cyclooxygenase-2 inhibitor blocks expression of mediators of renal injury in a model of diabetes and hypertension. Kidney Int 62:929 939 Braun L, Kardon T, Reisz-Porszasz ZS, Banhegyi G, Mandl J 2001 The regulation of the induction of vascular endothelial growth factor at the onset of diabetes in spontaneously diabetic rats. Life Sci 69:25332542 Hoshi S, Shu Y, Yoshida F, Inagaki T, Sonoda J, Watanabe T, Nomoto K, Nagata M 2002 Podocyte injury promotes progressive nephropathy in Zucker diabetic fatty rats. Lab Invest 82:2535 Abdel Aziz MY, Ben Gharbia O, el-Sayed Mohamed K, Muchaneta-Kubara EC, El Nahas AM 1997 VEGF and diabetic microvascular complications. Nephrol Dial Transplant 12:1538 Chiarelli F, Spagnoli A, Basciani F, Tumini S, Mezzetti A, Cipollone F, Cuccurullo F, Morgese G, Verrotti A 2000 Vascular endothelial growth factor (VEGF) in children, adolescents and young adults with type 1 diabetes mellitus: relation to glycaemic control and microvascular complications. Diabet Med 17:650 656 McLaren M, Elhadd TA, Greene SA, Belch JJ 1999 Elevated plasma vascular endothelial cell growth factor and thrombomodulin in juvenile diabetic patients. Clin Appl Thromb Hemost 5:2124 Diamant M, Hanemaaijer R, Verheijen JH, Smit JW, Radder JK, Lemkes HH 2001 Elevated matrix metalloproteinase-2 and -9 in urine, but not in serum, are markers of type 1 diabetic nephropathy. Diabet Med 18:423 424 Malamitsi-Puchner A, Sarandakou A, Tziotis J, Dafogianni C, Bartsocas CS 1998 Serum levels of basic fibroblast growth factor and vascular endothelial growth factor in children and adolescents with type 1 diabetes mellitus. Pediatr Res 44:873 875 Hovind P, Tarnow L, Oestergaard PB, Parving H-H 2000 Elevated vascular endothelial growth factor in type 1 diabetic patients with diabetic nephropathy. Kidney Int Suppl 75:S56 S61 Chaturvedi N, Fuller JH, Pokras F, Rottiers R, Papazoglou N, Aiello LP 2001 Circulating plasma vascular endothelial growth factor and microvascular complications of type 1 diabetes mellitus: the influence of ACE inhibition. Diabet Med 18:288 294 Brausewetter F, Jehle PM, Jung MF, Boehm BO, Brueckel J, Hombach V, Osterhues HH 2001 Microvascular permeability is increased in both types of diabetes and correlates differentially with serum levels of insulin-like growth factor I (IGF-I) and vascular endothelial growth factor (VEGF). Horm Metab Res 33:713720 Honkanen EO, Teppo AM, Gronhagen-Riska C 2000 Decreased urinary excretion of vascular endothelial growth factor in idiopathic membranous glomerulonephritis. Kidney Int 57:23432349 Gro ne HJ, Simon M, Gro ne EF 1995 Expression of vascular endothelial growth factor in renal vascular disease and renal allografts. J Pathol 177:259 267 Shulman K, Rosen S, Tognazzi K, Manseau EJ, Brown LF 1996 Expression of vascular permeability factor (VPF/VEGF) is altered in many glomerular diseases. J Am Soc Nephrol 7:661 666 Wasada T, Kawahara R, Katsumori K, Naruse M, Omori Y 1998 Plasma concentration of immunoreactive vascular endothelial growth factor and its relation to smoking. Metabolism 47:2730 Shimada K, Baba T, Neugebauer S, Onozaki A, Yamada D, Midorikawa S, Sato W, Watanabe T 2002 Plasma vascular endothelial growth factor in Japanese type 2 diabetic patients with and without nephropathy. J Diabetes Complications 16:386 390 Bortoloso E, Del Prete D, Gambaro G, Dalla Vestra M, Sailer A, Baggio B, Antonucci F, Fioretto P, Anglani F 2001 Vascular endothelial growth factor (VEGF) and VEGF receptors in diabetic nephropathy: expression studies in biopsies of type 2 diabetic patients. Ren Fail 23:483 493 Flyvbjerg A, Dagnaes-Hansen F, De Vriese AS, Schrijvers BF,

Endocrine Reviews, December 2004, 25(6):9711010 1009 Tilton RG, Rasch R 2002 Amelioration of long-term renal changes in obese type 2 diabetic mice by a neutralizing vascular endothelial growth factor antibody. Diabetes 51:3090 3094 Schrijvers BF, Rasch R, Tilton RG, Flyvbjerg A 2002 High proteininduced glomerular hypertrophy is vascular endothelial growth factor-dependent. Kidney Int 61:1600 1604 Flyvbjerg A, Schrijvers BF, De Vriese AS, Tilton RG, Rasch R 2002 Compensatory glomerular growth after unilateral nephrectomy is VEGF dependent. Am J Physiol Endocrinol Metab 283: E362E366 Fong TA, Shawver LK, Sun L, Tang C, App H, Powell TJ, Kim YH, Schreck R, Wang X, Risau W, Ullrich A, Hirth KP, McMahon G 1999 SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth of multiple tumor types. Cancer Res 59:99 106 Gilbert RE, Kelly DJ, Cox AJ, Wilkinson-Berka JL, Rumble JR, Osicka T, Panagiotopoulos S, Lee V, Hendrich EC, Jerums G, Cooper ME 2000 Angiotensin converting enzyme inhibition reduces retinal overexpression of vascular endothelial growth factor and hyperpermeability in experimental diabetes. Diabetologia 43: 1360 1367 Heldin CH, Westermark B 1999 Mechanism of action and in vivo role of platelet-derived growth factor. Physiol Rev 79:12831316 Changsirikulchai S, Hudkins KL, Goodpaster TA, Volpone J, Topouzis S, Gilbertson DG, Alpers CE 2002 Localization of platelet derived growth factor-D (PDGF-D) and its receptor in human kidney. Kidney Int 62:20432054 Gilbertson DG, Duff ME, West JW, Kelly JD, Sheppard PO, Hofstrand PD, Gao Z, Shoemaker K, Bukowski TR, Moore M, Feldhaus AL, Humes JM, Palmer TE, Hart CE 2001 Plateletderived growth factor C (PDGF-C), a novel growth factor that binds to PDGF and receptor. J Biol Chem 276:27406 27414 Shultz PJ, DiCorleto PE, Silver BJ, Abboud HE 1988 Mesangial cells express PDGF mRNAs and proliferate in response to PDGF. Am J Physiol 255:F674 F684 Alpers CE, Hudkins KL, Ferguson M, Johnson RJ, Rutledge JC 1995 Platelet-derived growth factor A-chain expression in developing and mature human kidneys and in Wilms tumor. Kidney Int 48:146 154 Seifert RA, Alpers CE, Bowen-Pope DF 1998 Expression of platelet-derived growth factor and its receptors in the developing and adult mouse kidney. Kidney Int 54:731746 Alpers CE, Seifert RA, Hudkins KL, Johnson RJ, Bowen-Pope DF 1992 Developmental patterns of PDGF B-chain, PDGF-receptor, and -actin expression in human glomerulogenesis. Kidney Int 42:390 399 Floege J, Johnson RJ, Alpers CE, Fatemi-Nainie S, Richardson CA, Gordon K, Couser WG 1993 Visceral glomerular epithelial cells can proliferate in vivo and synthesize platelet-derived growth factor B-chain. Am J Pathol 142:637 650 Nakagawa H, Sasahara M, Haneda M, Koya D, Hazama F, Kikkawa R 2000 Immunohistochemical characterization of glomerular PDGF B-chain and PDGF -receptor expression in diabetic rats. Diabetes Res Clin Pract 48:8798 Eitner F, Ostendorf T, Van Roeyen C, Kitahara M, Li X, Aase K, Grone HJ, Eriksson U, Floege J 2002 Expression of a novel PDGF isoform, PDGF-C, in normal and diseased rat kidney. J Am Soc Nephrol 13:910 917 Floege J, Hudkins KL, Seifert RA, Francki A, Bowen-Pope DF, Alpers CE 1997 Localization of PDGF -receptor in the developing and mature human kidney. Kidney Int 51:1140 1150 Gesualdo L, Di Paolo S, Milani S, Pinzani M, Grappone C, Ranieri E, Pannarale G, Schena FP 1994 Expression of plateletderived growth factor receptors in normal and diseased human kidney. An immunohistochemistry and in situ hybridization study. J Clin Invest 94:50 58 Di Paolo S, Gesualdo L, Ranieri E, Grandaliano G, Schena FP 1996 High glucose concentration induces the overexpression of transforming growth factor- through the activation of a plateletderived growth factor loop in human mesangial cells. Am J Pathol 149:20952106 Jones SC, Saunders HJ, Qi W, Pollock CA 1999 Intermittent high

422. 423.

442. 443.

424.

444.

425.

445.

426. 427. 428.

446. 447.

429. 430.

448.

449. 450.

431.

432. 433.

451. 452.

434.

453.

435. 436. 437. 438. 439.

454.

455.

456. 457.

440.

458.

441.

459.

Downloaded from edrv.endojournals.org on April 19, 2006

1010

Endocrine Reviews, December 2004, 25(6):9711010

Schrijvers et al. Factors Influencing Diabetic Nephropathy 469. Lev-Ran A, Hwang DL 1990 Epidermal growth factor and plateletderived growth factor in blood in diabetes mellitus. Acta Endocrinol (Copenh) 123:326 330 470. Fagerudd JA, Groop PH, Honkanen E, Teppo AM, Gro nhagenRiska C 1997 Urinary excretion of TGF-1, PDGF-BB and fibronectin in insulin- dependent diabetes mellitus patients. Kidney Int Suppl 63:S195S197 471. Guillausseau PJ, Dupuy E, Bryckaert MC, Timsit J, Chanson P, Tobelem G, Caen JP, Lubetzki J 1989 Platelet-derived growth factor (PDGF) in type 1 diabetes mellitus. Eur J Clin Invest 19: 172175 472. Kanauchi M, Nishioka M, Dohi K 2000 Secreted protein acidic and rich in cysteine (SPARC) in patients with diabetic nephropathy and tubulointerstitial injury. Diabetologia 43:1076 1077 473. Langham RG, Kelly DJ, Maguire J, Dowling JP, Gilbert RE, Thomson NM 2003 Over-expression of platelet-derived growth factor in human diabetic nephropathy. Nephrol Dial Transplant 18:13921396 474. Gilbert RE, Kelly DJ, McKay T, Chadban S, Hill PA, Cooper ME, Atkins RC, Nikolic-Paterson DJ 2001 PDGF signal transduction inhibition ameliorates experimental mesangial proliferative glomerulonephritis. Kidney Int 59:1324 1332 475. Grandaliano G, Ranieri E, Monno R, Gesualdo L, Schena F 1999 Ramipril inhibits in vitro human mesangial cell proliferation and platelet-derived growth factor expression. Exp Nephrol 7:229 235 476. Fukui M, Nakamura T, Ebihara I, Makita Y, Osada S, Tomino Y, Koide H 1994 Effects of enalapril on endothelin-1 and growth factor gene expression in diabetic rat glomeruli. J Lab Clin Med 123: 763768 477. Kohno M, Ikeda M, Johchi M, Horio T, Yasunari K, Kurihara N, Takeda T 1993 Interaction of PDGF and natriuretic peptides on mesangial cell proliferation and endothelin secretion. Am J Physiol 265:E673E679 478. Grandaliano G, Biswas P, Choudhury GG, Abboud HE 1993 Simvastatin inhibits PDGF-induced DNA synthesis in human glomerular mesangial cells. Kidney Int 44:503508 479. Gesualdo L, Di Paolo S, Ranieri E, Schena FP 1994 Trapidil inhibits human mesangial cell proliferation: effect on PDGF -receptor binding and expression. Kidney Int 46:10021009

460.

461.

462.

463.

464.

465. 466.

467. 468.

glucose enhances cell growth and collagen synthesis in cultured human tubulointerstitial cells. Diabetologia 42:11131119 Inaba T, Ishibashi S, Gotoda T, Kawamura M, Morino N, Nojima Y, Kawakami M, Yazaki Y, Yamada N 1996 Enhanced expression of platelet-derived growth factor- receptor by high glucose. Involvement of platelet-derived growth factor in diabetic angiopathy. Diabetes 45:507512 Fraser D, Wakefield L, Phillips A 2002 Independent regulation of transforming growth factor-1 transcription and translation by glucose and platelet-derived growth factor. Am J Pathol 161:1039 1049 Throckmorton DC, Brogden AP, Min B, Rasmussen H, Kashgarian M 1995 PDGF and TGF- mediate collagen production by mesangial cells exposed to advanced glycosylation end products. Kidney Int 48:111117 Pichler RH, Bassuk JA, Hugo C, Reed MJ, Eng E, Gordon KL, Pippin J, Alpers CE, Couser WG, Sage EH, Johnson RJ 1996 SPARC is expressed by mesangial cells in experimental mesangial proliferative nephritis and inhibits platelet-derived-growth-factormedicated mesangial cell proliferation in vitro. Am J Pathol 148: 11531167 Choudhury GG, Karamitsos C, Hernandez J, Gentilini A, Bardgette J, Abboud HE 1997 PI-3-kinase and MAPK regulate mesangial cell proliferation and migration in response to PDGF. Am J Physiol 273:F931F938 Young BA, Johnson RJ, Alpers CE, Eng E, Gordon K, Floege J, Couser WG, Seidel K 1995 Cellular events in the evolution of experimental diabetic nephropathy. Kidney Int 47:935944 Kelly DJ, Gilbert RE, Cox AJ, Soulis T, Jerums G, Cooper ME 2001 Aminoguanidine ameliorates overexpression of prosclerotic growth factors and collagen deposition in experimental diabetic nephropathy. J Am Soc Nephrol 12:2098 2107 Gilbert RE, McNally PG, Cox A, Dziadek M, Rumble J, Cooper ME, Jerums G 1995 SPARC gene expression is reduced in early diabetes-related kidney growth. Kidney Int 48:1216 1225 Riley SG, Steadman R, Williams JD, Floege J, Phillips AO 1999 Augmentation of kidney injury by basic fibroblast growth factor or platelet-derived growth factor does not induce progressive diabetic nephropathy in the Goto Kakizaki model of non-insulin-dependent diabetes. J Lab Clin Med 134:304 312

Endocrine Reviews is published bimonthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.

Downloaded from edrv.endojournals.org on April 19, 2006

Вам также может понравиться