Skip to main content

Main menu

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Subject Collections
    • JASN Podcasts
    • Archives
    • Saved Searches
    • ASN Meeting Abstracts
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Subscriptions
  • More
    • About JASN
    • Alerts
    • Advertising
    • Editorial Fellowship Program
    • Feedback
    • Reprints
    • Impact Factor
  • ASN Kidney News
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
American Society of Nephrology
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart
Advertisement
American Society of Nephrology

Advanced Search

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Subject Collections
    • JASN Podcasts
    • Archives
    • Saved Searches
    • ASN Meeting Abstracts
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Subscriptions
  • More
    • About JASN
    • Alerts
    • Advertising
    • Editorial Fellowship Program
    • Feedback
    • Reprints
    • Impact Factor
  • ASN Kidney News
  • Follow JASN on Twitter
  • Visit ASN on Facebook
  • Follow JASN on RSS
  • Community Forum
FRONTIERS IN NEPHROLOGY
You have accessRestricted Access

Extracellular Matrix Metabolism in Diabetic Nephropathy

Roger M. Mason and Nadia Abdel Wahab
JASN May 2003, 14 (5) 1358-1373; DOI: https://doi.org/10.1097/01.ASN.0000065640.77499.D7
Roger M. Mason
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nadia Abdel Wahab
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data Supps
  • Info & Metrics
  • View PDF
Loading

Abstract

ABSTRACT. Diabetic nephropathy is characterized by excessive deposition of extracellular matrix proteins in the mesangium and basement membrane of the glomerulus and in the renal tubulointerstitium. This review summarizes the main changes in protein composition of the glomerular mesangium and basement membrane and the evidence that, in the mesangium, these are initiated by changes in glucose metabolism and the formation of advanced glycation end products. Both processes generate reactive oxygen species (ROS). The review includes discussion of how ROS may activate intracellular signaling pathways leading to the activation of redox-sensitive transcription factors. This in turn leads to change in the expression of genes encoding extracellular matrix proteins and the protease systems responsible for their turnover. E-mail: roger.mason@imperial.ac.uk

Diabetic Nephropathy: The Magnitude of the Problem

Diabetes mellitus has recently assumed epidemic proportions, partly due to the 2 to 5% increase in the incidence of type 1 diabetes in children, but especially due to the global increase in type 2 diabetes (1). Currently as many as 4% of obese adolescents in the United States may have silent type 2 diabetes (2). Secondary microvascular complications, including nephropathy, develop some years after the onset of diabetes. Genetic background is likely to be important in determining susceptibility to diabetic nephropathy (DN) (3), but exposure of tissues to chronic hyperglycaemia is the main initiating factor (4,5⇓). The prevalence of nephropathy varies according to geographical location, type of diabetes, and the length of time since diagnosis. Microalbuminuria is a sign of early DN, while macroalbuminuria indicates progression (6). While there is a similar prevalence of microalbuminuria in diabetic patients in the Unite States and Europe (22 and 25%), the prevalence of macroalbuminuria is higher in the United States (27% compared with 12%) (7). In a Japanese study, 44% of type 2 diabetic patients developed DN by 30 yr after diagnosis but only 20% of type 1 patients did so (8). Despite these regional variations, the prevalence of DN is predicted to increase in the decades ahead (9,10⇓). DN is a major cause of end-stage renal disease (11), and new therapeutic approaches are required to limit its development. Evolution of these will depend on understanding the molecular mechanisms driving glomerulosclerosis and interstitial fibrosis in DN. This review surveys current understanding in this area.

Changes in Renal Extracellular Matrices in DN

Similar ultrastructural changes occur in glomeruli in types 1 and 2 diabetes (12,13⇓). The glomerular basement membrane increases in thickness, and the extracellular matrix of the mesangium expands. The basement membrane changes, accompanied by glomerular hyperfiltration, and increased glomerular hydrostatic pressure lead to microalbuminuria. However, the mesangial changes appear to be the main cause of declining renal function in DN (14). As the mesangial matrix expands, it impinges on glomerular capillaries, reducing the surface available for filtration and narrowing or occluding the lumen. Declining glomerular function correlates well with the extent of these changes in both types of diabetes (15,16⇓), although there appears to be more heterogeneity in these structural changes in type 2 (17).

Tubulointerstitial fibrosis occurs in DN, in addition to glomerulosclerosis, but it has been much less well studied. However, decreased creatinine clearance correlates with the interstitial expansion as well as with mesangial expansion (18) and survival rates diminish if interstitial fibrosis is present (19). In this respect, diabetic nephropathy is similar to other renal disorders in which progressive loss of renal function correlates with advancing interstitial fibrosis (20). Interstitial fibrosis in DN may be initiated by the same factors as glomerular fibrosis, as well as being influenced subsequently by factors originating in the glomerulus (21).

Expansion of the mesangial matrix and thickening of the glomerular basement membrane (GBM) in DN could be due to either increased accumulation of proteins that are normally present in these structures or to deposition of proteins that are not present in normal tissue or to both. Table 1 summarizes some of the many reports of matrix proteins that are present in the mesangium and GBM in DN. It is clear that some mesangial proteins such as collagen I and III are only expressed in the late stages of glomerulosclerosis. They are associated with the development of Kimmelstiel-Wilson nodules rather than with the diffuse expansion of the mesangial matrix, which occurs in the early and moderately advanced stages of the disease. Other proteins such as fibronectin are present in the normal mesangium but increase in the expanding mesangium.

View this table:
  • View inline
  • View popup

Table 1. Glomerular matrix proteins in diabetic nephropathy (DN)

Differences between quantitative electron microscopy-immunogold measurements of the levels of matrix proteins in the mesangium and GBM and data obtained by immunohistochemical and immunofluorescence studies complicates the interpretation of which proteins are responsible for expansion or thickening. Thus an immunogold analysis showed decreased type VI collagen per unit area of the mesangium and decreased total type VI per glomerulus mesangial matrix in “fast track” DN patients with diffuse mesangial expansion compared with controls (30). In contrast, immunohistochemical and immunofluorescence investigations concluded that increased type VI deposition occurs (25,32⇓). Such differences could be due to the immunogold study focusing on an earlier stage of the disease and on type 1 patients, whereas the light microscopy studies were largely concerned with late-stage DN in type 2 diabetic patients. Nevertheless, the quantitative study provokes the question as to precisely which matrix proteins are responsible for diffuse mesangial expansion and whether there may be subtle differences in the molecular changes in type 1 and 2 diabetes. Only further quantitative investigations on well-defined cohorts of patients will provide definitive answers.

The main components of the normal GBM are type IV collagen, laminin, entactin, and proteoglycans. GBM type IV collagen is comprised predominantly of α3, α4, and α5 chains (39,40⇓). Table 1 summarizes some of the changes that occur during thickening of the GBM in DN. The loss of heparan sulfate proteoglycans with disease progression is associated with proteinuria (44,46⇓), the glycosaminoglycan chains normally forming an anionic charge barrier to protein diffusion across the GBM (49).

Genetically determined diabetes mellitus occurs in db/db mice (a model for type 2), and type 1 diabetes can be induced in mice and rats with streptozotocin (STZ). These models develop nephropathy with changes in glomerular extracellular proteins that appear to be similar to those in human DN. There is increased glomerular expression and accumulation of type IV collagen and fibronectin (50–53⇓⇓⇓), while the relative content of heparan sulfate proteoglycan content is reduced (50). As in human DN, hyperglycemia is likely to be the main factor initiating these changes.

Studies using in situ hybridization to detect specific mRNAs indicate that increased expression of several different genes encoding matrix proteins is likely to be responsible, in part, for their accumulation in glomerular matrices in DN in both humans (24,26,54⇓⇓) and in animal models (51,53⇓). However, glomerular matrix proteins also normally undergo metabolic turnover, being degraded primarily by matrix metalloproteinases (MMP) (55). Any decrease in turnover would promote excessive matrix accumulation, and there is evidence that this too occurs in DN. Glomerular mRNA levels for MMP2 (gelatinase A) and MMP3 (stromelysin) decrease in human DN (54,56⇓), while in STZ-induced diabetes in rats, MMP9 (gelatinase B) mRNA and activity fell and tissue inhibitor of metalloproteinase-1 (TIMP1) mRNA increased (57,58⇓). In this model, MMP3 mRNA levels increased (58). Nevertheless, MMP3 activity and overall glomerular MMP activity decreased (58,59⇓). Activity of MMP is controlled at multiple steps, including their level of synthesis as inactive proenzymes, activation of the proenzymes and, subsequently, inhibition of active enzymes by binding to TIMP (60). Plasmin cleaves and activates MMP proenzymes, while membrane type metalloproteinase-1 (MT1-MMP) is involved specifically in activating MMP2. In vitro evidence indicates that high-glucose conditions reduce plasmin activity by upregulating expression of plasminogen activator inhibitor (PAI-1) in mesangial cells (MC) (61,62⇓) while suppressing expression of MT1-MMP (63). Thus, overall, turnover of mesangial matrix proteins in DN is likely to be compromised by decreased MMP expression, decreased levels of proenzyme activation, and increased expression of the MMP inhibitor, TIMP1.

Mesangial Cell Responses to Hyperglycemic Conditions In Vitro

Many in vitro investigations have exposed MC to high concentrations of glucose, or to albumin-Amadori adducts, or to albumin-advanced glycation end products (AGE) to mimic the conditions they experience in vivo. The formation of Amadori-protein adducts and AGE-proteins is initiated in vivo by the non-enzymatic interaction of protein amino-groups with glucose (64) or with metabolites of glucose such as methylglyoxal (65). Increased levels of glycated proteins occur in the plasma, renal, and other tissues in diabetes (66–68⇓⇓). MC respond to culture medium containing high glucose concentration or glycated-albumin by upregulating the expression of many of the matrix proteins that accumulate in the mesangium in DN, and by changes in the expression of MMP and proteinase inhibitors. Some examples are shown in Table 2. It is apparent that both induce similar effects, even though those of glucose depend on its metabolism, following uptake into the cell by facilitative glucose transporters (75), while glycated-proteins interact with cell surface receptors (76,77⇓).

View this table:
  • View inline
  • View popup

Table 2. Mesangial cell protein expression in vitro in response to hyperglycemic stimulia

As well as being exposed to circulating glycated-proteins, mesangial and other cells are likely to be affected by the function of intracellular proteins being compromised by AGE formation and by cellular interactions with insoluble glycated matrix proteins. The latter lead to some of the same effects in vitro as are induced by high glucose; for example, increased expression of MMP2 and TIMP1 and decreased expression of MT1-MMP (78). Glycation of type IV collagen and laminin affects their structure and assembly into polymers (79–81⇓⇓). Formation of advanced glycation end products of collagen IV or glomerular basement membrane also markedly inhibits their susceptibility to cleavage by MMP3 and MMP9 (82), impeding their turnover.

Other glomerular cells also respond to high-glucose conditions or to glycated-albumin in vitro. For example, glomerular visceral epithelial cells exposed to high glucose have been reported to upregulate expression of transforming growth factor-β (TGF-β) and fibronectin (83) and TGF-β type II receptor (84), while glycated-albumin stimulates the production of collagen IV and fibronectin in glomerular endothelial cells (85).

The response of MC to hyperglycemia is clearly complex, and differential gene screening techniques have proved useful for understanding more fully the changes in gene expression that occur after exposure to high glucose. mRNA differential display indicated that a large number of genes undergo changes in expression after exposure of human MC to 30 mM D-glucose (86), and suppression subtraction hybridization (SSH) subsequently identified 70 known genes that were induced under these conditions, a further 26 showing similarity to expressed sequence tags (EST) and 100 cDNAs representing genes that are downregulated (87,88⇓).

Differential screening of MC exposed to high glucose revealed increased expression of genes that, potentially, play a role in the development of glomerulosclerosis, including connective tissue growth factor (CTGF) (87,89⇓). Its role in promoting the synthesis of mesangial matrix proteins is discussed below. However, expression screening also indicated that mRNA for both subunits of the fibronectin receptor (α5,β1 integrin) is increased in high-glucose conditions (N.A. Wahab and R.M. Mason, unpublished work). Further experiments demonstrated that TGF-β, acting at least in part through a CTGF-dependent pathway, increases the number of α5β1 receptors on the cell surface and their occupation by ligand (90). This promotes increased deposition of insoluble fibronectin matrix around the cells, a process that can be inhibited by anti-β1-neutralizing antibody (90). This mechanism may represent one mechanism for the development of fibrosis in the mesangium in DN. It is also conceivable that an increased number of mesangial cell surface α5β1 integrin sites ligated by fibronectin may modulate cell behavior by an outside-in signaling mechanism. MC cultured in three-dimensional collagen gels show fibronectin-dependent activation of phosphsatidylinositol-4-phosphate 5-kinase by outside-in signaling (91). Fibronectin signaling stimulates protein kinase C (PKC) translocation to the cell membrane in CHO cells (92) and induces hypertrophy of cardiac myocytes, accompanied by increased transcription of the brain natriuretic peptide gene (93).

Changes in Intermediary Metabolism in Response to Hyperglycemia

A high plasma glucose concentration leads to elevated intracellular glucose levels in cells such as MC in which uptake is not regulated by insulin. The increased uptake in MC is probably achieved by increased expression of GLUT1 in response to high glucose. GLUT 1 is a high affinity, low capacity facilitative glucose transporter which is near saturated at normal physiologic concentrations of glucose (75). Glucose is normally metabolized through glycolysis and the tricarboxylic acid cycle, generating CO2 and the reduced coenzymes NADH and FADH2. The latter reoxidize by donating electrons to the respiratory chain (electron transport chain) in the inner mitochondrial membrane, where they reduce molecular oxygen to form water. Passage of electrons through the chain results in a proton gradient across the membrane which drives oxidative phosphorylation, producing ATP. High intracellular glucose levels generate increased production of several glucose metabolites which are normally present in only low concentration and the glucose and three of these products stimulate activity of four other pathways, and actions downstream of them. Thus (1) diacylglycerol (DAG), derived from the glycolytic intermediate dihydroxacetone phosphate (DHAP), activates several isoforms of PKC; (2) dicarbonyl metabolites such as glyoxal and methylglyoxal, derived from glucose and the glycolytic intermediates DHAP and glyceraldehyde-3-phosphate (G3P), initiate AGE formation; (3) fructose-6-phosphate, a glycolytic intermediate, when present in raised concentration, enters and increases flux through the hexosamine biosynthetic pathway, generating UDP-N-acetylglucosamine. This promotes glycosylation of Sp1 and possibly of other transcription factors, modulating their activity; (4) glucose itself, which when present in raised concentration enters the aldose reductase pathway and is converted to sorbitol. This utilizes the reduced coenzyme NADPH. The resulting NADP is converted back to NADPH using the cell’s antioxidant, reduced glutathione, depleting the latter and increasing cell susceptibility to reactive oxygen species (ROS). The details of activation of these four pathways in high glucose have been reviewed extensively recently (94,95⇓). Increased production of NADH and FADH2 and donation of electrons from them to the respiratory chain increases the proton gradient across the inner mitochondrial membrane, which inhibits electron transport at complex III of the chain. Electrons carried by coenzyme Q, which normally pass to complex III, then generate intracellular ROS by reducing O2 to superoxide ion O2− (94). This also inhibits activity of the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (96), potentially raising the level of metabolic intermediates between glucose and G3P and their entry into the four pathways. The formation of AGE also generates extracellular ROS due to the autooxidation of glucose (97). This may lead to the activation of latent TGFβ (L-TGFβ) (98). Extracellular AGE also bind to the cell surface receptor, RAGE, which is expressed in various renal cell types (76,77⇓). This initiates intracellular ROS production (99).

Attenuation of DN in Animal Models of Diabetes

Support for the involvement of ROS and AGE in inducing DN in vivo comes from studies on the effect of various inhibitors on animal models. Thus, overexpression of Cu2+/Zn2+ superoxide dismutase in mice protects them from glomerular injury after induction of diabetes with STZ (100). Inhibitors of non-enzymatic glycation of proteins attenuate glomerular and interstitial damage in db/db mice (101–106⇓⇓⇓⇓⇓) and in diabetic transgenic mice overexpressing RAGE (a receptor for AGE), which otherwise develop advanced glomerulosclerosis (107). Likewise, inhibition of PKCβ attenuates mesangial expansion in db/db mice (52) and glomerular dysfunction in the STZ rat (108). Sorbinil, an inhibitor of the aldose reductase pathway, reduced GBM width in STZ rats on low-protein diets but not in those on higher-protein diets (109). However, definitive evidence of the efficacy of antioxidants, or inhibitors of AGE-formation, PKC activation, or of the aldose reductase pathways, in attenuating the development of DN in humans is still awaited (95).

To date, there are no in vivo studies on the effect of blocking the hexosamine biosynthetic pathway on glomerular function in diabetic models. However, inhibition of glutamine:fructose-6-phosphate-amidotransferase (GFAT), the rate-limiting enzyme of the pathway, inhibits high-glucose–induced TGF-β production (110) and NF-κB-dependent promoter activation (111), two key events in the response to high glucose (see below). Overexpression of GFAT activates the PAI-1 promoter (112), another key event.

Growth Factors and Hormones Affecting Matrix Protein Expression in DN

In vitro and in vivo studies have shown that several different growth factors are upregulated in DN, and the signaling pathways these activate ultimately regulate transcription factors affecting glomerular ECM accumulation. The principle factors and hormones involved are angiotensin II (AngII), TGF-β, CTGF, platelet derived growth factor (PDGF), growth hormone (GH), and insulin-like growth factors (IGF).

Angiotensin II.

Activation of the renal renin-angiotensin system (RAS) and its involvement in the pathogenesis of DN is indicated by several studies that showed that both angiotensin-converting enzyme (ACE) inhibitors and angiotensin-receptor-I antagonists attenuate DN (11,113–114⇓⇓). The entire RAS is present in the kidney (115). In vitro studies show increased angiotensin expression in mesangial and tubular cells in response to glucose (116,117⇓). This response was shown to be mediated, at least in part, by ROS and subsequent activation of p38 MAPK in tubular cells (118). AngII, the active octapeptide derived from angiotensinogen, also stimulates ROS generation in vascular smooth muscle cells (119) and MC (120). The ROS signaling pathway activates NF-κB, which mediates further angiotensinogen expression, so ROS generation could be part of a positive feedback loop (121), perpetuating activity of the RAS in DN. Increased AngII is also generated in hypertension, a disorder that frequently accompanies diabetes and accelerates progression of DN (122). Several intrarenal hemodynamic changes, including glomerular capillary hypertension, occur in early experimental diabetes. This and the subsequent development of albuminuria and glomerular structural changes is attenuated by ACE inhibition (123), demonstrating the importance of hemodynamic factors in the pathogenesis of DN. It is noteworthy that intraglomerular hypertension can occur in diabetes even in the absence of systemic hypertension (124). Although AngII is associated with both systemic and renal hemodynamic effects, it also has direct nonhemodynamic effects on glomerular cells, particularly MC. AngII has been shown to increase ECM accumulation by MC, primarily via stimulation of TGF-β expression (53,115,125–127⇓⇓⇓⇓). The mechanism by which AngII transactivates TGF-β was found to be similar to that for hyperglycemia. Both transactivate the growth factor gene through two AP-1 regulatory elements (128–130⇓⇓). This activation appears to be PKC- and p38 MAPK-dependent (129,130⇓).

Transforming Growth Factor-β.

Numerous studies indicate that hyperglycemia induces an increase in TGF-β expression on both the mRNA and protein levels in experimental and human diabetes (51,127,130–132⇓⇓⇓⇓) as well as in cultured MC (71,133,134⇓⇓). TGF-β appears to be involved in both the early and later stages of DN. In STZ-diabetic rats, renal TGF-β expression increased markedly as early as 24 h after the onset of hyperglycemia (135). A similar increase was also reported in the non-obese diabetic (NOD) mouse (136,137⇓) and in the diabetic BB rat (136). Sustained elevated expression of TGF-β occurs in STZ-diabetic rats with diabetes of 24-wk duration (138) and in the kidney of 16-wk-old diabetic db/db mice (139). All TGF-β isoforms, TGF-β1, β2, and β3, and the TGF-β type II receptor were reported to be elevated in experimental diabetes (139–141⇓⇓). Increased renal production of TGF-β in patients with diabetes and DN is also well documented (127,142,143⇓⇓). In addition, many in vitro studies have shown that TGF-β expression is increased in various renal cells cultured under high-glucose conditions or with AGE (71,73,143–145⇓⇓⇓⇓).

It is now clear that TGF-β is the key cytokine mediating the production of different ECM proteins in MC, epithelial cells, renal interstitial cells, and fibroblasts (71,133,144,146–151⇓⇓⇓⇓⇓⇓⇓⇓). TGF-β also influences matrix-degrading enzymes by inhibiting the synthesis of collagenases and stimulating the production of TIMP and PAI-1 (149,150,152–154⇓⇓⇓⇓). TGF-β can also influence local matrix deposition by upregulating different ECM receptors (90,150⇓).

Although the main signaling pathway of TGF-β is the Smad pathway (155) and is activated in the STZ-diabetic mouse model (156), TGF-β also activates other pathways such as the MAPK (ERK, SAPK/JNK, and p38 MAPK) (157,158⇓). TGF-β induces PKA activation in MC by a mechanism involving the degradation of the inhibitory peptide of PKA (PKI) (159).

Connective Tissue Growth Factor.

CTGF is another prosclerotic cytokine and has also been shown to be involved in both the early and later stages of DN. Its expression is increased in experimental diabetic glomerulosclerosis (160,161⇓). Elevated CTGF levels in glomeruli of NOD mice appear to correlate with the duration of diabetes (89). Elevated CTGF expression was also detected in human DN (89,162,163⇓⇓). In vitro studies have shown that CTGF is induced in renal cells by both high glucose and AGE (87,89,160⇓⇓), as well as ROS (164), which they generate. The induction of CTGF in MC by hyperglycemia seems to be partly TGF-β–dependent and partly PKC-dependent (165,166⇓).

CTGF is one of the TGF-β-inducible immediate early genes (167) and is induced in cultured MC by TGF-β (168–170⇓⇓). TGF-β induces CTGF gene expression via Smad binding elements (SBE) and a unique TGF-β response element in the CTGF promoter (170–172⇓⇓). The induction of CTGF by TGF-β seems to be PKC- and MAPK-dependent (170).

Emerging evidence from in vitro studies of renal cells, including MC, indicates that CTGF is a crucial mediator for TGF-β-stimulated matrix protein expression. CTGF has been shown to mediate TGF-β-induced increases in fibronectin (89,168,173⇓⇓), collagen type I (174–176⇓⇓), and the fibronectin receptor (α5β1 integrin) (90).

Other ECM proteins that have been reported to be induced by CTGF in different renal cells include collagens I, III, and IV, tenascin, and thrombospondin-1 (TSP-1) (87,177,178⇓⇓). Induction of FN by CTGF in MC appears to be mediated by the activation of MAPK and PKB pathways (179). Recent work in our laboratory has shown that MC exposed to CTGF downregulate expression of Smad7 (Wahab and Mason, unpublished work). Smad7 is an inhibitor of the Smad signaling pathway (155); we therefore propose that CTGF promotes increased TGF-β signaling through this pathway due to decreased availability of Smad7. We hypothesize that this accounts, at least in part, for CTGF’s profibrotic activity.

Platelet-Derived Growth Factor.

Glomerular mRNA levels for PDGF-B chain are enhanced fivefold in experimental diabetes (106). PDGF has been reported to mediate both high glucose– and AGE-dependent induction of type III collagen in cultured rat MC. However, its effect seems to be through augmenting the production of TGF-β (180,181⇓). Anti-PDGF antibodies also attenuated the increased production of type IV collagen in MC exposed to AGE, again suggesting that PDGF acts as an intermediate factor (182).

Growth Hormone and Insulin-Like Growth Factors.

Classically, GH secreted from the pituitary induces the synthesis of IGF in various tissues through activation of the GH receptor (GHR). Two lines of evidence implicate GH in the pathogenesis of glomerular fibrosis in experimental diabetes. One is the renoprotective effect of long-acting somatostatin analogs and GHR antagonists (183). The other is the failure of STZ-treated mice, which are either transgenic for a mutated GH, which is an antagonist of native GH, or in which the GHR/binding protein gene has been knocked out, to develop glomerular lesions (184–186⇓⇓). Transgenic mice that overexpress GH or GH releasing factor develop glomerulosclerosis, but those expressing an IGF-1 transgene have morphologically normal, though enlarged, glomeruli (187). Overall the evidence favors GH having a direct effect in the pathogenesis of glomerulosclerosis, independent of IGF. Plasma GH levels are raised in type I diabetic patients with poor glycemic control, while the concentration of IGF-I is low and that of IGF binding protein 1 (IGFBP1), a modulator of its activity, is raised (e.g., 188). Intraportal insulin deficiency decreases hepatic IGF-I synthesis and increases production of IGFBP in type I diabetes, while hypersecretion of GH occurs as a result of lowered negative feedback of IGF-1 on secretion (189).

How GH induces glomerulosclerosis directly is not known, but mRNA for GHR has been detected in MC and they respond to the hormone in vitro by upregulating iNOS transcripts and nitric oxide production (190). However, iNOS knockout mice show more advanced mesangial expansion, increased GBM staining for collagen IV, and tubulointerstitial fibrosis, after induction of diabetes with STZ than do controls, suggesting a protective effect for NO in vivo with respect to matrix deposition (191). On binding its cellular receptor, GH activates the JAK/STAT, MAPK, and P13K signaling pathways (192), so it may potentially regulate the transcription of a number of genes.

IGF-I may be expressed in the kidney independently of GH. MC cultured on glycated-albumin increase IGF-I production (193), and the growth factor directly stimulates synthesis of laminin, fibronectin, proteoglycan, and type IV collagen in these cells (194–196⇓⇓). IGF-I and IGF-BP species have also been reported to increase in the kidney in the early stages of experimental DN (197–200⇓⇓⇓), while the expression level of IGF-IR rises in a later phase (201). IGF-I and IGF-II signal through two specific receptors, IGF-IR and IGFII/mannose-6-phosphate receptor (IGF-II/man-6-PR) (202,203⇓). IGF-I activates the phosphatidylinositol-3-kinase (PI3K) and ERK1/2 MAPK pathways (196,204⇓). It has also been shown to stimulate NF-κB (205). In the presence of high glucose, IGF increased IGF-I-stimulated insulin receptor substrate-1/2 phosphorylation and AP-1 transcriptional activity, while decreasing IGFBP-2 expression (204). Thus, despite lower circulating levels of IGF-I in diabetes, locally produced growth factor may influence MC in vivo. MC from diabetic NOD mice secrete increased amounts of IGF-I, and this probably contributes to the increased ECM accumulation, largely through an IGF-I-mediated reduction in MMP2 activity (206).

ROS and the Activation of Pathways Stimulating Matrix Accumulation

Although further in vitro and in vivo confirmatory studies are required, current data strongly suggest that intracellular ROS, from either AGE-RAGE interaction or glucose metabolism (94,207⇓), have a direct role in overproduction of ECM proteins and that this can be counteracted by antioxidants (208). It has been shown that ROS activate the PKC, MAPK, and JAK-STAT pathways (209–212⇓⇓⇓), which lead to the activation of redox-sensitive transcription factors including NF-κB, AP-1 (Fos and Jun proteins), STAT, and Egr-1 (99,213–215⇓⇓⇓). These enhance the transactivation of genes coding for cytokines such as TGF-β and CTGF that upregulate ECM protein expression (164,216–217⇓⇓).

Other Factors Modulating the Activity of TGF-β

Thombospondin-1.

TSP-1 is one of five isoforms of thrombospondin and is synthesized and secreted by a variety of renal cells, including MC (86,218⇓). Exposure to high glucose upregulates TSP-1 expression in MC (86), and increased levels of glycoprotein occur in the glomeruli in diabetic animals (219) and in DN in man (220). In addition to interacting with many other matrix proteins (221,222⇓), TSP-1 also modulates their level of synthesis (223). This appears to be primarily through its ability to activate latent TGF-β (224). In vitro studies indicate that TSP-1–dependent TGF-β1 activation is important in the mesangial cell response to high glucose (225,226⇓). Interestingly, TSP-1 null mice have a similar phenotype to TGF-β null mice (227), suggesting an important role for the TSP-1 activation mechanism in vivo.

Decorin.

The expression of decorin (DCN), a small proteoglycan containing a single dermatan/chondroitin sulfate chain, is markedly upregulated in MC exposed to high glucose (71,228⇓) and in the glomerulus in DN, although protein accumulation only occurs in the late stage of the disease (24). Decorin binds to collagen type I (229) and to other extracellular proteins, including activated TGF-β, which it sequesters in the matrix (230), possibly as a ternary complex with fibrillar collagen in advancing DN (24). Decorin-TGF-β complexes are also excreted in the urine in late DN (24). Administration of DCN inhibits TGF-β–mediated ECM expression in experimental nephritis (230). We recently showed that DCN inhibits TGF-β–mediated upregulation of PAI-1 in MC through a mechanism that involves Ca2+-dependent phosphorylation of Smad2 at a key regulatory site, thus modulating the TGF-β Smad signaling pathway (231). Despite all these studies the role of decorin in DN remains unresolved.

Hyperglycemia and the Regulation of Gene Expression

Any hypothesis explaining the overall coordinated program of changes in gene expression in hyperglycemia that lead to glomerulosclerosis must take into account the activation mechanism of the genes involved. An E-box has been implicated as a carbohydrate response element in TGF-β and several other glucose-regulated promoters (232,233⇓). However, the concept of a single glucose response element being implicated in the overall changes in gene expression becomes less attractive when considering the very large number of genes upregulated by hyperglycemia. Table 3 summarizes promoter region analyses for a number of these human genes and includes known hyperglycemia-associated stimuli, the response elements, transcription factors, and signaling pathways activating them.

View this table:
  • View inline
  • View popup

Table 3. Activation of genes encoding matrix proteins in hyperglycemic conditionsa

We propose that ROS are the overall activators of these signaling pathways (Figure 1). Thus matrix-associated latent TGF-β is activated by ROS generated via extracellular formation of AGE and interaction of the active factor with its receptor activates the Smad signaling pathway. ROS generated intracellularly from glucose metabolism and AGE-RAGE interaction activates PKC (together with DAG) and MAPK pathways. This leads to the rapid activation of cellular “redox-sensitive” transcription factors such as NF-κB, AP-1 (fos and jun proteins), Erg-1, and Stat1. These, together with activated Smads (phosphorylated Smad2 and 3 complexed with Smad4), coordinate the transcription of a wave of genes, including angiotensinogen, TSP-1, and CTGF. AngII derived from angiotensinogen stimulates further generation of ROS and expression of TGF-β. Secreted CTGF works in concert with TGF-β activated by TSP-1 and ROS to transactivate subsequent waves of genes, including those encoding structural proteins whose accumulation leads to glomerulosclerosis in DN (Figure 1).

Figure1
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 1. Model to explain the mechanisms by which hyperglycemia promotes ECM protein accumulation by mesangial cells. Hyperglycemic conditions generate reactive oxygen species (ROS), which activate a cascade of events. Downstream of these, TGF-β and connective tissue growth factor (CTGF) work in a coordinated manner to promote increased expression of ECM proteins. See text for details.

It is becoming clear that the coordinated expression of TGF-β and CTGF is crucial for the induction of ECM proteins and thus for the development of DN. The expression of some ECM proteins, such as fibronectin, is CTGF-dependent, and its promoter region does not contain any Smad binding elements (SBE) (89). It is also noteworthy that previous experiments have shown that TGF-β induces fibronectin expression via a MAPK-dependent pathway and not via a Smad-dependent pathway (250), although a recent report indicates that a Smad-dependent pathway may operate in mice (251). In contrast, the transcription of a number of other matrix proteins, for example collagen I, PAI-1, and TIMP-1, is TGF-β-dependent, and their promoter regions contain SBE (Table 3). However, it appears that increased signaling by TGF-β is also markedly influenced by CTGF, as we have found that the latter rapidly inhibits the expression level of Smad7. This inhibitory Smad mediates an intracellular negative feedback that limits TGF-β signaling (Figure 1), apparently by blocking the association of R-Smads (Smad2 and 3) with the TGF-β receptor complex and, thereby, their phosphorylation (252). Overexpression of Smad7 has been shown to block TGF-β–dependent induction of collagen I in MC (253). Thus CTGF may mediate the induction of ECM protein expression both directly and indirectly by potentiating the TGF-β/Smad signaling pathway. A signaling receptor for CTGF has not yet been characterized, but rapid phosphorylation of MAPK after exposure to the growth factor indicates that one must be present in responsive cells (179,254⇓).

The promoters of MMP genes also show remarkable conservation of regulatory elements (255), including AP-1, ETS, and the TGF-β inhibitory element, TIE. The downregulation of MMP3 (256), collagenase, MMP9, and MMP7 (257) is thought to be mediated by TIE. There is also evidence for modulation of MMP7 mRNA stability through ATTTA motifs in the 3′-untranslated region (258). With respect to this we have shown that high-glucose conditions switch off synthesis of a protein, HGRG-14, in MC (259). This is achieved by production in high glucose of an HGRG-14 mRNA with a long 3′ UTR containing several destabilizing ATTTA motifs, which has a short half-life and is not translated. In low glucose, an mRNA with a short 3′ UTR and no ATTTA motifs is produced that has a longer half life and is translated (259).

Concluding Comments

The response of glomerular cells, especially MC, to hyperglycemia, is driven primarily by the generation of ROS and then TGF-β and CTGF. This upregulates the transcription of many matrix genes and represses that of MMP, events which in vivo lead to glomerulosclerosis (Figure 1). It is likely that the response of other renal cells to hyperglycemia has many features in common with this pathway, but this has been less well studied to date. There are clearly many points at which therapeutic approaches could be tried to provide renoprotection in diabetes. These include ACE inhibitors and AT-1 receptor antagonists, which are already in use clinically, and novel agents to oppose the actions of ROS, TSP-1, TGF-β, and CTGF. It is likely that targeting multiple points in altered metabolism in the diabetic kidney will be more successful in attenuating the development of DN, due to its complexity, rather than a single approach.

Acknowledgments

We are grateful for financial support from the Medical Research Council UK and Diabetes UK. We thank Linda Readings for secretarial support.

  • © 2003 American Society of Nephrology

References

  1. ↵
    Silink M: Childhood diabetes: A global perspective. Horm Res 57 [Suppl 1]: 1–5, 2002
    OpenUrl
  2. ↵
    Sinha R, Fisch G, Teague B, Tamorlane WV, Banyas B, Allen K, Savoye M, Rieger V, Taksali S, Barbetta G, Sherwin RS, Caprio S: Prevalence of impaired glucose tolerance among children and adolescents with marked obesity. N Engl J Med 346: 802–810, 2002
    OpenUrlCrossRefPubMed
  3. ↵
    Quinn M, Angelico MC, Warram JH, Krolewski AS: Familial factors determine the development of diabetic nephropathy in patients with IDDM. Diabetologia 39: 940–945, 1996
    OpenUrlCrossRefPubMed
  4. ↵
    The Diabetes Control and Complications Trial Research Group: The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 329: 977–986, 1993
    OpenUrlCrossRefPubMed
  5. ↵
    UK Prospective Diabetes Study (UKPDS) Group: Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 352: 837–853, 1998
    OpenUrlCrossRefPubMed
  6. ↵
    Mogensen CE, Christensen CK, Vittinghus E: The stages in diabetic renal disease. With emphasis on the stage of incipient diabetic nephropathy. Diabetes, 32 [Suppl 2]: 64–78, 1983
  7. ↵
    Lloyd CE, Stephenson J, Fuller JH, Orchard TJ: A comparison of renal disease across two continents: The epidemiology of diabetes complications study and the EURODIAB IDDM Complications Study. Diabetes Care 19: 219–225, 1996
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Yokoyama H, Okudaira M, Otani T, Sato A, Miura J, Takaike H, Yamada H, Muto K, Uchigata Y, Ohashi Y, Iwamoto Y: Higher incidence of diabetic nephropathy in type 2 than in type 1 diabetes in early-onset diabetes in Japan. Kidney Int 58: 302–311, 2000
    OpenUrlCrossRefPubMed
  9. ↵
    Green A, Sjolie AK, Eshoj O: Trends in the epidemiology of IDDM during 1970–2020 in Fyn County. Denmark. Diabetes Care 19: 801–806, 1996
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Bagust A, Hopkinson PK, Maslove L, Currie CJ: The projected health care burden of type 2 diabetes in the UK from 2000 to 2060. Diabetes Med 19 [Suppl 4]: 1–5, 2002
    OpenUrl
  11. ↵
    Ritz E, Orth SR: Nephropathy in patients with type 2 diabetes mellitus. N Engl J Med 341: 1127–1133, 1999
    OpenUrlCrossRefPubMed
  12. ↵
    Osterby R: Glomerular structural changes in type 1 (insulin-dependent) diabetes mellitus: Causes, consequences and prevention. Diabetologia 35: 803–812, 1992
    OpenUrlCrossRefPubMed
  13. ↵
    Osterby R, Gall MA, Schmitz A, Nielsen FS, Nyberg G, Parving HH: Glomerular structure and function in proteinuric type 2 (non-insulin-dependent) diabetic patients. Diabetologia 36: 1064–1070, 1993
    OpenUrlCrossRefPubMed
  14. ↵
    Steffes MW, Osterby R, Chavers B, Mauer SM: Mesangial expansion as a central mechanism for loss of kidney function in diabetic patients. Diabetes 38: 1077–1081, 1989
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Osterby R, Tapia J, Nyberg G, Tencer J, Willner J, Rippe B, Torffvit O: Renal structures in type 2 diabetic patients with elevated albumin excretion rate. APMIS 109: 751–761, 2001
    OpenUrlCrossRefPubMed
  16. ↵
    Osterby R, Hartmann A, Nyengaard JR, Bangstad HJ: Development of renal structural lesions in type 1 diabetic patients with microalbuminuria. Observations by light microscopy in 8-year follow-up biopsies. Virchows Arch 440: 94–101, 2002
    OpenUrlCrossRefPubMed
  17. ↵
    Dalla Vestra M, Saller A, Bortoloso E, Mauer M, Fioretto P: Structural involvement in type 1 and type 2 diabetic nephropathy. Diabetes Metab 26 [Suppl 4]: 8–14, 2000
  18. ↵
    White KE, Bilous RW: Type 2 diabetic patients with nephropathy show structural-functional relationships that are similar to type 1 disease. J Am Soc Nephrol 11: 1667–1673, 2000
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Bohle A, Wehrmann M, Bogenschutz O, Batz C, Muller CA, Muller GA: The pathogenesis of chronic renal failure in diabetic nephropathy. Investigation of 488 cases of diabetic glomerulosclerosis. Pathol Res Pract 187: 251–259, 1991
    OpenUrlCrossRefPubMed
  20. ↵
    Risdon RA, Sloper JC, de Wardener HE: Relationship between renal function and histological changes found in renal-biopsy specimens from patients with persistent glomerular nephritis. Lancet 2 7564: 363–366, 1968
    OpenUrl
  21. ↵
    Gilbert RE, Cooper ME: The tubulointerstitium in progressive diabetic kidney disease: More than an aftermath of glomerular injury? Kidney Int 56: 1627–1637, 1999
    OpenUrlCrossRefPubMed
  22. Glick AD, Jacobson HR, Haralson MA: Mesangial deposition of type I collagen in human glomerulosclerosis. Hum Pathol 23: 1373–1379, 1992
    OpenUrlCrossRefPubMed
  23. Stokes MB, Holler S, Cui Y, Hudkins KL, Eitner F, Fogo A, Alpers, CE: Expression of decorin, biglycan and collagen type I in human renal fibrosing disease. Kidney Int 57: 487–498, 2000
    OpenUrlCrossRefPubMed
  24. ↵
    Schaefer L, Raslik I, Grone HJ, Schonherr E, Macakova K, Ugorcakova J, Budny S, Schaefer RM, Kresse H: Small proteoglycans in human diabetic nephropathy: discrepancy between glomerular expression and protein accumulation of decorin, biglycan, lumican, and fibromodulin. FASEB J 15: 559–561, 2001
    OpenUrlCrossRefPubMed
  25. ↵
    Makino H, Shikata K, Wieslander J, Wada J, Kashinara N, Yoshioka K, Ota Z: Localization of fibril/microfibril and basement membrane collagens in diabetic glomerulosclerosis in type 2 diabetes. Diabet Med 11: 304–311, 1994
    OpenUrlPubMed
  26. ↵
    Razzaque MS, Koji T, Taguchi T, Harada T, Namane PK: In situ localization of type III and type IV collagen-expressing cells in human diabetic nephropathy. J Pathol 174: 131–138, 1994
    OpenUrlCrossRefPubMed
  27. Makino H, Kashihara N, Sugiyama H, Kanao K, Sekikawa T, Shikata K, Nagai R, Ota Z: Phenotypic changes of the mesangium in diabetic nephropathy. J Diabetes Complications 9: 282–284, 1995
    OpenUrlCrossRefPubMed
  28. Kim Y, Kleppel MM, Butkowski R, Mauer SM, Wieslander J, Michael AF: Differential expression of basement membrane collagen chains in diabetic nephropathy. Am J Pathol 138: 413–420, 1991
    OpenUrlPubMed
  29. Makino H, Shikata K, Hayashi T, Wieslander J, Haramoto T, Hirata K, Wada J, Yoshida T, Yoshioka K, Ota Z: Immunoreactivity of the JK-132 monoclonal antibody directed against basement membrane collagen in normal and diabetic glomeruli. Virchows Arch 434: 235–241, 1994
    OpenUrlCrossRef
  30. ↵
    Zhu D, Kim Y, Steffes MW, Groppoli TJ, Butkowski RJ, Mauer SM: Application of electron microscopic immunocytochemistry to the human kidney: Distribution of type IV and type VI collagen in normal human kidney. J Histochem Cytochem 42: 577–584, 1994
    OpenUrlCrossRefPubMed
  31. Yagame M, Kim Y, Zhu D, Suzuki D, Eguchi K, Nomoto Y, Sakai H, Groppoli T, Steffes MW, Mauer SM: Differential distribution of type IV collagen chains in patients with diabetic nephropathy in non-insulin-dependent diabetes mellitus. Nephron 70: 42–48, 1995
    OpenUrlCrossRefPubMed
  32. ↵
    Nerlich A, Schleicher E: Immunohistochemical localization of extracellular matrix components in human diabetic glomerular lesions. Am J Pathol 139: 889–899, 1991
    OpenUrlPubMed
  33. Kiryu K, Morita H, Fujita Y, Kawasumi M, Shinzato T, Tsuruta Y, Nakai S, Maeda K: Phenotypic expressions of type I III, IV, V and VI collagens in patients with diabetic nephropathy: Immunohistochemical comparison between HD and non-HD patients. Nippon Jinzo Gakkai Shi 36: 365–373, 1994
    OpenUrlPubMed
  34. Razzaque MS, Koji T, Harada T, Taguchi T: Identification of type VI collagen synthesizing cells in human diabetic glomerulosclerosis using renal biopsy sections. Anal Cell Pathol 15: 175–181, 1997
    OpenUrlPubMed
  35. Moriya T, Groppoli TJ, Kim Y, Mauer M: Quantitative immunoelectron microscopy of type VI collagen in glomeruli in type I diabetic patients. Kidney Int 59: 317–323, 2001
    OpenUrlCrossRefPubMed
  36. Makino H, Ikeda S, Haramoto T, Ota Z: Heparan sulfate proteoglycans are lost in patients with diabetic nephropathy. Nephron 61: 415–421, 1992
    OpenUrlCrossRefPubMed
  37. Suzuki D: Measurement of the extracellular matrix in glomeruli from patients with diabetic nephropathy using an automatic image analyser. Nippon Jinzo Gakkai Shi 36: 1209–1215, 1994
    OpenUrlPubMed
  38. Van Vliet A, Baelde HJ, Vleming LJ, de Heer E, Bruijn JA: Distribution of fibronectin isoforms in human renal disease. J Pathol 193: 256–262, 2001
    OpenUrlCrossRefPubMed
  39. ↵
    Zeisbert M, Ericksen MB, Hamano Y, Neilson EG, Ziyadeh F, Kalluri R: Differential expression of type IV collagen isoforms in rat glomerular endothelial and mesangial cells. Biochem Biophys Res Commun 295: 401–407, 2002
    OpenUrlCrossRefPubMed
  40. ↵
    Borza DB, Bondar O, Todd P, Sundaramoorthy M, Sado Y, Ninomiya Y, Hudson BG: Quaternary organization of the goodpasture autoantigen, the α3(IV) collagen chain. Sequestration of two cryptic autoepitopes by intrapromoter interactions with the α4 and α5 NC1 domains. J Biol Chem 277: 40075–40083, 2002
    OpenUrlAbstract/FREE Full Text
  41. Zhu D, Kim Y, Steffes MW, Groppoli TJ, Butkowski RJ, Mauer SM: Glomerular distribution of type IV collagen in diabetes by high resolution quantitative immunochemistry. Kidney Int 45: 425–433, 1994
    OpenUrlPubMed
  42. Katz A, Fish AJ, Kleppel MM, Hagen SG, Michael AF, Butkowski RJ: Renal entactin (nidogen): Isolation, characterization and tissue distribution. Kidney Int 40: 643–652, 1991
    OpenUrlPubMed
  43. Falk RJ, Scheinman JI, Mauer SM, Michael AF: Polyantigenic expansion of basement membrane constituents in diabetic nephropathy. Diabetes 32 [Suppl 2]: 34–39, 1983
  44. ↵
    Ikeda S, Makino H, Haramoto T, Shikata K, Kumagai I, Ota Z: Changes in glomerular extracellular matrices components in diabetic nephropathy. J Diabet Complications 5: 186–188, 1991
    OpenUrlCrossRefPubMed
  45. Shimomura H, Spiro RG: Studies on macromolecular components of human glomerular basement membrane and alterations in diabetes. Decreased levels of heparan sulfate proteoglycans and laminin. Diabetes 36: 374–381, 1987
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Vernier RL, Steffes MW, Sisson-Ross S, Mauer SM: Heparan sulfate proteoglycans in the glomerular basement membrane in type 1 diabetes mellitus. Kidney Int 41: 1070–1080, 1992
    OpenUrlCrossRefPubMed
  47. Makino H, Yamasaki Y, Haramoto T, Shikata K, Hironaka K, Ota Z, Kanwar YS: Ultrastructural changes of extracellular matrices in diabetic nephropathy revealed by high resolution scanning and immunoelectron microscopy. Lab Invest 68: 45–55, 1993
    OpenUrlPubMed
  48. Tamsma JT, van den Born J, Bruijn JA, Assmann KJ, Weening JJ, Berden JH, Wieslander J, Schrama E, Hermans J, Veerkamp JH, et al: Expression of glomerular extracellular matrix components in human diabetic nephropathy: Decrease of heparan sulphate in the glomerular basement membrane. Diabetologia 37: 313–320, 1994
    OpenUrlCrossRefPubMed
  49. ↵
    Kanwar YS, Linker A, Farquhar MG: Increased permeability of the glomerular basement membrane to ferritin after removal of glycosaminoglycans (heparan sulfate) by enzyme digestion. J Cell Biol 86: 688–693, 1980
    OpenUrlAbstract/FREE Full Text
  50. ↵
    van den Born J, van Kraats AA, Bakker MA, Assmann KJ, van den Heuvel LP, Veerkamp JH, Berden JH: Selective proteinuria in diabetic nephropathy in the rat is associated with a relative decrease in glomerular basement membrane heparan sulphate. Diabetologia 38: 161–172, 1995
    OpenUrlPubMed
  51. ↵
    Park IS, Kiyomoto H, Abboud SL, Abboud HE: Expression of transforming growth factor-β and type IV collagen in early streptozotocin-induced diabetes. Diabetes 46: 473–480, 1997
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, King GL, Kikkawa R: Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC-β inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J 14: 439–447, 2000
    OpenUrlCrossRefPubMed
  53. ↵
    Ziyadeh FN, Hoffman BB, Han DC, Iglesias-De La Cruz MC, Hong SW, Isono M, Chen S, McGowan TA, Sharma K: Long-term prevention of renal insufficiency, excess matrix gene expression, and glomerular mesangial matrix expansion by treatment with monoclonal anti-transforming growth factor-β antibody in db/db diabetic mice. Proc Natl Acad Sci USA 97: 8015–8020, 2000
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Suzuki D, Miyazaki M, Jinde K, Koji T, Yagame M, Endo M, Nomoto Y, Sakai H: In situ hybridization studies of matrix metalloproteinase-3, tissue inhibitor of metalloproteniase-1 and type IV collagen in diabetic nephropathy. Kidney Int 52: 111–119, 1997
    OpenUrlCrossRefPubMed
  55. ↵
    Davies M, Coles GA, Thomas GJ, Martin J, Lovett DH: Proteinases and the glomerulus: Their role in glomerular diseases. Klin Wochenschr 68: 1145–1149, 1990
    OpenUrlCrossRefPubMed
  56. ↵
    Del Prete D, Anglani F, Forino M, Ceol M, Fioretto P, Nosadini R, Baggio B, Gambaro G: Down-regulation of glomerular matrix metalloproteinase-2 gene in human NIDDM. Diabetologia 40: 1449–1454, 1997
    OpenUrlCrossRefPubMed
  57. ↵
    Shankland SJ, Ly H, Thai K, Scholey JW: Glomerular expression of tissue inhibitor of metalloproteinase (TIMP-1) in normal and diabetic rats. J Am Soc Nephrol 7: 97–104, 1996
    OpenUrlAbstract
  58. ↵
    McLennan SV, Kelly DJ, Cox AJ, Cao Z, Lyons JG, Yue DK, Gilbert RE: Decreased matrix degradation in diabetic nephropathy: Effects of ACE inhibition on the expression and activities of matrix metalloproteinases. Diabetologia 45: 268–275, 2002
    OpenUrlCrossRefPubMed
  59. ↵
    Reckelhoff JF, Tygart VL, Mitias MM, Walcott JL: STZ-induced diabetes results in decreased activity of glomerular cathepsin and metalloprotease in rats. Diabetes 42: 1425–1432, 1993
    OpenUrlAbstract/FREE Full Text
  60. ↵
    Sternlicht MD, Werb Z: How matrix metalloproteinases regulate cell behaviour. Annu Rev Cell Dev Biol 17: 463–516, 2001
    OpenUrlCrossRefPubMed
  61. ↵
    Abdel Wahab NA, Mason RM: Modulation of neutral protease expression in human mesangial cells by hyperglycaemic culture. Biochem J 320: 777–783, 1996
  62. ↵
    Fisher EJ, McLennan SV, Yue DK, Turtle JR: High glucose reduces generation of plasmin activity by mesangial cells. Microvasc Res 53: 173–178, 1997
    OpenUrlCrossRefPubMed
  63. ↵
    McLennan SV, Martell SY, Yue DK: High glucose concentration inhibits the expression of membrane type metalloproteinase by mesangial cells: Possible role in mesangium accumulation. Diabetologia 43: 642–648, 2000
    OpenUrlCrossRefPubMed
  64. ↵
    Vlassara H, Brownlee M, Cerami A: Nonenzymatic glycosylation: Role in the pathogenesis of diabetic complications. Clin Chem 32: B37–B41, 1986
  65. ↵
    Degenhardt TP, Thorpe SR, Baynes JW: Chemical modification of proteins by methylglyoxal. Cell Mol Biol 44: 1139–1145, 1998
    OpenUrlPubMed
  66. ↵
    Bucala R, Vlassara H: Advanced glycosylation end products in diabetic renal and vascular disease. Am J Kidney Dis 26: 875–888, 1995
    OpenUrlCrossRefPubMed
  67. ↵
    Sakai H, Jinde K, Suzuki D, Yagame M, Nomoto Y: Localization of glycated proteins in the glomeruli of patients with diabetic nephropathy. Nephrol Dial Transplant 11 [Suppl 5]: 66–71, 1996
  68. ↵
    Horie K, Miyata T, Maeda K, Miyata S, Sugiyama S, Sakai H, van Ypersole de Strihou C, Monnier VM, Witztum JL, Kurokawa K: Immunohistochemical colocalization of glycoxidation products and lipid peroxidation products in diabetic renal glomerular lesions. Implications for glycoxidative stress in the pathogenesis of diabetic nephropathy. J Clin Invest 100: 2995–3004, 1997
    OpenUrlCrossRefPubMed
  69. Ayo SH, Radnik RA, Garoni JA, Glass WF2nd, Kreisberg JI: High glucose causes an increase in extracellular matrix proteins in cultured mesangial cells. Am J Pathol 136: 1339–1348, 1990
    OpenUrlPubMed
  70. Haneda M, Kikkawa R, Horide N, Togawa M, Koya D, Kajiwara N, Ooshima A, Shigeta Y: Glucose enhances type IV collagen production in cultured rat glomerular mesangial cells. Diabetologia 34: 198–200, 1991
    OpenUrlCrossRefPubMed
  71. ↵
    Wahab NA, Harper K, Mason RM: Expression of extracellular matrix molecules in human mesangial cells in response to prolonged hyperglycaemia. Biochem J 316: 985–992, 1996
  72. Cohen MP, Hud E, Wy VY, Ziyadeh FN: Albumin modified by Amadori glucose adducts activates mesangial cell type IV collagen gene transcription. Mol Cell Biochem 151: 61–67, 1995
    OpenUrlCrossRefPubMed
  73. ↵
    Ziyadeh FN, Han DC, Cohen JA, Guo J, Cohen MP: Glycated albumin stimulates fibronectin gene expression in glomerular mesangial cells: Involvement of the transforming growth factor-beta system. Kidney Int 53: 631–638, 1998
    OpenUrlCrossRefPubMed
  74. Zheng F, Cai W, Mitsuhashi T, Vlassara H: Lysozyme enhances renal excretion of advanced glycation end products in vivo and suppresses adverse age-mediated cellular effects in vitro: a potential AGE sequestration therapy for diabetic nephropathy? Mol Med 7: 737–747, 2001
    OpenUrlPubMed
  75. ↵
    Heilig CW, Brosius FC3rd, Henry DN: Glucose transporters of the glomerulus and the implications for diabetic nephropathy. Kidney Int Suppl 60: S91–S99, 1997
    OpenUrlPubMed
  76. ↵
    Tanji N, Markowitz GS, Fu C, Kislinger T, Taguchi A, Pischetsrieder M, Stern D, Schmidt AM, D’Agati VD: 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, 2000
    OpenUrlAbstract/FREE Full Text
  77. ↵
    Vlassara H: The AGE-receptor in the pathogenesis of diabetic complications. Diabetes Metab Res Rev 17: 436–443, 2001
    OpenUrlCrossRefPubMed
  78. ↵
    McLennan SV, Martell SK, Yue DK: Effects of mesangium glycation on matrix metalloproteinase activities: Possible role in diabetic complications. Diabetes 51: 2612–2618, 2002
    OpenUrlAbstract/FREE Full Text
  79. ↵
    Tsilibary EC, Charonis AS, Reger LA, Wohlhueter RM, Furcht LT: The effect of nonenzymatic glucosylation on the binding of the main noncollagenous NC1 domain to type IV collagen. J Biol Chem 263: 4302–4308, 1988
    OpenUrlAbstract/FREE Full Text
  80. ↵
    Tanaka S, Avigad G, Brodsky B, Eikenberry EF: Glycation induces expansion of the molecular packing of collagen. J Mol Biol 203: 495–505, 1988
    OpenUrlCrossRefPubMed
  81. ↵
    Charonis AS, Tsilbary EC: Structural and functional changes of laminin and type IV collagen after nonenzymatic glycation. Diabetes 41 [Suppl 2]: 49–51, 1992
  82. ↵
    Mott JD, Khalifah RG, Nagase H, Shield CF3rd, Hudson JK, Hudson BG: Nonenzymatic glycation of type IV collagen and matrix metalloproteinase susceptibility. Kidney Int 52: 1302–1312, 1997
    OpenUrlCrossRefPubMed
  83. ↵
    van Det NF, Verhagen NA, Tamsma JT, Berden JH, Bruijn JA, Daha MR, van der Woude FJ: Regulation of glomerular epithelial cell production of fibronectin and transforming growth factor-β by high glucose, not by angiotensin II. Diabetes 46: 834–840, 1997
    OpenUrlAbstract/FREE Full Text
  84. ↵
    Iglesias-de la Cruz MC, Ziyadeh FN, Isono M, Kouahou M, Han DC, Kalluri R, Mundel P, Chen S: Effects of high glucose and TGF-β1 on the expression of collagen IV and vascular endothelial growth factor in mouse podocytes. Kidney Int 62: 901–913, 2002
    OpenUrlCrossRefPubMed
  85. ↵
    Cohen MP, Wu VY, Cohen JA: Glycated albumin stimulates fibronectin and collagen IV production by glomerular endothelial cells under normoglycemic conditions. Biochem Biophys Res Commun 239: 91–94, 1997
    OpenUrlCrossRefPubMed
  86. ↵
    Holmes DI, Abdel Wahab NA, Mason RM: Identification of glucose-regulated genes in human mesangial cells by mRNA differential display. Biochem Biophys Res Commun 238: 179–184, 1997
    OpenUrlCrossRefPubMed
  87. ↵
    Murphy M, Godson C, Cannon S, Kato S, Mackenzie HS, Martin F, Brady HR: Suppression subtractive hybridization identifies high glucose levels as a stimulus for expression of connective tissue growth factor and other genes in human mesangial cells. J Biol Chem 274: 5830–5834, 1999
    OpenUrlAbstract/FREE Full Text
  88. ↵
    Clarkson MR, Murphy M, Gupta S, Lambe T, Mackenzie HS, Godson C, Martin F, Brady HR: High glucose-altered gene expression in mesangial cells. Actin-regulatory protein gene expression is triggered by oxidative stress and cytoskeletal disassembly. J Biol Chem 277: 9707–9712, 2002
    OpenUrlAbstract/FREE Full Text
  89. ↵
    Wahab NA, Yevdokimova N, Weston BS, Roberts T, Li XJ, Brinkman H, Mason RM: Role of connective tissue growth factor in the pathogenesis of diabetic nephropathy. Biochem J 359: 77–87, 2001
    OpenUrlCrossRefPubMed
  90. ↵
    Weston BS, Wahab NA, Mason RM: CTGF mediates TGFβ-induced fibronectin matrix deposition by upregulating active α5β1 integrin in human mesangial cells. J Am Soc Nephrol 14: 601–610, 2003
    OpenUrlAbstract/FREE Full Text
  91. ↵
    Dunlop ME, Muggli EE: Extracellular matrix components cooperate to activate phosphatidyl inositol-4-phosphate 5-kinase. Biochem Biophys Res Commun 279: 931–937, 2000
    OpenUrlCrossRefPubMed
  92. ↵
    Kreuzer J, Schmidts A, Marquetant R, Niebauer J, Strasser RH: Protein kinase C activation after cellular adhesion on fibronectin: Partial suppression after inhibition of protein isoprenylation. Eur J Med Res 2: 305–310, 1997
    OpenUrlPubMed
  93. ↵
    Ogawa E, Saito Y, Kuwahara K, Harada M, Miyamoto Y, Hamanaka I, Kajiyama N, Takahashi N, Izumi T, Kawakami R, Kishimoto I, Naruse Y, Mori N, Nakao K: Fibronectin signaling stimulates BNP gene transcription by inhibiting neuron-restrictive silencer element-dependent repression. Cardiovasc Res 53: 451–459, 2002
    OpenUrlCrossRefPubMed
  94. ↵
    Brownlee M: Biochemistry and molecular cell biology of diabetic complications. Nature 414: 813–820, 2001
    OpenUrlCrossRefPubMed
  95. ↵
    Sheetz MJ, King GL: Molecular understanding of hyperglycemia’s adverse effects for diabetic complications. JAMA 288: 2579–2588, 2002
    OpenUrlCrossRefPubMed
  96. ↵
    Du XL, Edelstein D, Rossetti L, Fantus IG, Goldberg H, Ziyadeh F, Wu J, Brownlee M: Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc Natl Acad Sci USA 97: 12222–12226, 2000
    OpenUrlAbstract/FREE Full Text
  97. ↵
    Baynes JW, Thorpe SR: Role of oxidative stress in diabetic complications: a new perspective on an old paradigm. Diabetes 48: 1–9, 1999
    OpenUrlAbstract
  98. ↵
    Barcellos-Hoff MH, Dix TA: Redox-mediated activation of latent transforming growth factor-beta 1. Mol Endocrinol 10: 1077–1083, 1996
    OpenUrlCrossRefPubMed
  99. ↵
    Yan SD, Schmidt AM, Anderson GM, Zhang J, Brett J, Zou YS, Pinsky D, Stern D: Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J Biol Chem 269: 9889–9897, 1994
    OpenUrlAbstract/FREE Full Text
  100. ↵
    Craven PA, Melhem MF, Phillips SL, DeRubertis FR: Overexpression of Cu2+/Zn2+ superoxide dismutase protects against early diabetic glomerular injury in transgenic mice. Diabetes 50: 2114–2125, 2001
    OpenUrlAbstract/FREE Full Text
  101. ↵
    Cohen MP, Masson N, Hud E, Ziyadeh F, Han DC, Clements RS: Inhibiting albumin glycation ameliorates diabetic nephropathy in the db/db mouse. Exp Nephrol 8: 135–143, 2000
    OpenUrlCrossRefPubMed
  102. ↵
    Nicholls K, Mandel TE: Advanced glycosylation end-products in experimental murine diabetic nephropathy: Effect of islet isografting and of aminoguanidine. Lab Invest 60: 486–491, 1989
    OpenUrlPubMed
  103. ↵
    Soulis T, Cooper ME, Vranes D, Bucala R, Jerums G: Effects of Aminoguanidine in preventing experimental diabetic nephropathy are related to the duration of treatment. Kidney Int 50: 627–634, 1996
    OpenUrlCrossRefPubMed
  104. ↵
    Tsuchida K, Makita Z, Yamagishi S, Atsumi T, Miyoshi H, Obara S, Ishida M, Ishikawa S, Yasamura K, Koike T: Suppression of transforming growth factor beta and vascular endothelial growth factor in diabetic nephropathy in rats by a novel advanced glycation end product inhibitor, OPB-9195. Diabetologia 42: 579–588, 1999
    OpenUrlCrossRefPubMed
  105. ↵
    Forbes JM, Soulis T, Thallas V, Panagiotopoulos S, Long DM, Vasan S, Wagle D, Jerums G, Cooper ME: Renoprotective effects of a novel inhibitor of advanced glycation. Diabetologia 44: 108–114, 2001
    OpenUrlCrossRefPubMed
  106. ↵
    Kelly DJ, Gilbert RE, Cox AJ, Soulis T, Jerums G, Cooper ME: Aminoguanidine ameliorates overexpression of prosclerotic growth factors and collagen deposition in experimental diabetic nephropathy. J Am Soc Nephrol 12: 2098–2107, 2001
    OpenUrlAbstract/FREE Full Text
  107. ↵
    Yamamoto Y, Kato I, Doi T, Yonekura H, Ohashi S, Takeuchi M, Watanabe T, Yamagishi S, Sakurai S, Takasawa S, Okamoto H, Yamamoto H: Development and prevention of advanced diabetic nephropathy in RAGE-overexpressing mice. J Clin Invest 108: 261–268, 2001
    OpenUrlCrossRefPubMed
  108. ↵
    Ishii H, Jirousek MR, Koya D, Takagi C, Xia P, Clermont A, Bursell SE, Kern TS, Ballas LM, Heath WF, Stramm LE, Feener EP, King GL: Amelioration of vascular dysfunction in diabetic rats by an oral PKC beta inhibitor. Science 272: 728–731, 1996
    OpenUrlAbstract/FREE Full Text
  109. ↵
    Mauer SM, Steffes MW, Azar S, Brown DM: Effects of sorbinil on glomerular structure and function in long-term diabetic rats. Diabetes 38: 839–846, 1989
    OpenUrlAbstract/FREE Full Text
  110. ↵
    Schleicher ED, Weigert C: Role of the hexosamine biosynthetic pathway in diabetic nephropathy. Kidney Int Suppl 77: S13–18, 2000
    OpenUrlCrossRefPubMed
  111. ↵
    James LR, Tang D, Ingram A, Ly H, Thai K, Cai L, Scholey JW: Flux through the hexosamine pathway is a determinant of nuclear factor kappaB-dependent promoter activation. Diabetes 51: 1146–1156, 2002
    OpenUrlAbstract/FREE Full Text
  112. ↵
    James LR, Fantus IG, Goldberg H, Ly H, Scholey JW: Overexpression of GFAT activates PAI-1 promoter in mesangial cells. Am J Physiol Renal Physiol 279: F718–F727, 2000
    OpenUrlPubMed
  113. ↵
    Lewis EJ, Hunsicker LG, Bain RP, Rohde RD: The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med 329: 1456–142, 1993
    OpenUrlCrossRefPubMed
  114. ↵
    Remuzzi A, Perico N, Amuchastegui CS, Malanchini B, Mazerska M, Battaglia C, Bertani T, Remuzzi G: Short- and long-term effect of angiotensin II receptor blockade in rats with experimental diabetes. J Am Soc Nephrol 4: 40–49, 1993
    OpenUrlAbstract
  115. ↵
    Wolf G, Ziyadeh FN: The role of angiotensin II in diabetic nephropathy: Emphasis on nonhemodynamic mechanisms. Am J Kidney Dis 29: 153–163, 1997
    OpenUrlCrossRefPubMed
  116. ↵
    Singh R, Alavi N, Singh AK, Leehey DJ: Role of angiotensin II in glucose-induced inhibition of mesangial matrix degradation. Diabetes 48: 2066–2073, 1999
    OpenUrlAbstract
  117. ↵
    Zhang SL, Filep JG, Hohman TC, Tang SS, Ingelfinger JR, Chan JSD: High levels of glucose stimulate angiotensinogen gene expression via the P38 mitogen-activated protein kinase pathway in rat kidney proximal tubular cells. Kidney Int 55: 454–464, 1999
    OpenUrlCrossRefPubMed
  118. ↵
    Hsieh TJ, Zhang SL, Filep JG, Tang SS, Ingelfinger JR, Chan JS: High glucose stimulates angiotensin gene expression via reactive oxygen species generation in rat kidney proximal tubular cells. Endocrinology 143: 2975–2985, 2002
    OpenUrlCrossRefPubMed
  119. ↵
    Ushio-Fukai-M, Zarari AM, Fukui T, Ishizaka N, Griendling KK: p22phox is a critical component of the superoxide-generating NADH/NADPH oxidase system and regulates angiotensin II-induced hypertrophy in vascular smooth muscle cells. J Biol Chem 271: 23317–23321, 1996
    OpenUrlAbstract/FREE Full Text
  120. ↵
    Jaimes EA, Galceran JM, Raij L: Angiotensin II induces superoxide anion production by mesangial cells. Kidney Int 54: 775–784, 1998
    OpenUrlCrossRefPubMed
  121. ↵
    Brasier AR, Jamaluddin M, Han Y, Patterson C, Runge MS: Angiotensin II induces gene transcription through cell-type-dependent effects on the nuclear factor-κB (NF-κB) transcription factor. Mol Cell Biochem 212: 155–196, 2000
    OpenUrlCrossRefPubMed
  122. ↵
    Jandeleit-Dahm K, Cooper ME: Hypertension and diabetes. Curr Opin Nephrol Hypertens 11: 221–228, 2002
    OpenUrlCrossRefPubMed
  123. ↵
    Zatz R, Dunn BR, Meyer TW, Anderson S, Rennke HG, Brenner BM: Prevention of diabetic glomerulopathy by pharmacological amelioration of glomerular capillary hypertension. J Clin Invest 77: 1925–1930, 1986
  124. ↵
    Cooper ME: Interaction of metabolic and haemodynamic factors in mediating experimental diabetic nephropathy. Diabetologia 44: 1957–1972, 2001
    OpenUrlCrossRefPubMed
  125. ↵
    Kagami S, Border WA, Miller DE, Noble NA: Angiotensin II stimulates extracellular matrix protein synthesis through induction of transforming growth factor-β expression in rat glomerular mesangial cells. J Clin Invest 93: 2431–2437, 1994
  126. ↵
    Wolf G: Link between angiotensin II and TGF-β in the kidney. Miner Electrolyte Metab 24: 174–180, 1998
    OpenUrlCrossRefPubMed
  127. ↵
    Yamamoto T, Noble NA, Cohen AH, Nast CC, Hishida A, Gold LI, Border WA: Expression of transforming growth factor-β isoforms in human glomerular diseases. Kidney Int 49: 461–469, 1996
    OpenUrlCrossRefPubMed
  128. ↵
    Kim SJ, Glick A, Sporn MB, Roberts AB: Characterization of the promoter region of the human transforming growth factor-β1 gene. J Biol Chem 264: 402–408, 1989
    OpenUrlAbstract/FREE Full Text
  129. ↵
    Weigert C, Sauer U, Brodbeck K, Pfeiffer A, Haring HU, Schleicher ED: AP-1 proteins mediate hyperglycemia-induced activation of the human TGF-β1 promoter in mesangial cells. J Am Soc Nephrol 11: 2007–1016, 2000
    OpenUrlAbstract/FREE Full Text
  130. ↵
    Weigert C, Brodbeck K, Klopfer K, Haring HU, Schleicher ED: Angiotensin II induces human TGF-β1 promoter activation: similarity to hyperglycemia. Diabetologia 45: 89–898, 2002
  131. ↵
    Sharma K and Ziyadeh FN: Hyperglycemia and diabetic kidney disease. The case for transforming growth factor-β as a key mediator. Diabetes 44: 439–446, 1995
    OpenUrl
  132. ↵
    Schleicher E, Nerlich A: The role of hyperglycemia in the development of diabetic complications. Horm Metab Res 28: 367–373, 1996
    OpenUrlCrossRefPubMed
  133. ↵
    Kolm V, Sauer U, Olgemooller B, Schleicher ED: High glucose-induced TGF-β1 regulates mesangial production of heparan sulfate proteoglycan Am J Physiol 270: F812–F821, 1996
  134. ↵
    Kolm-Litty V, Sauer U, Nerlich A, Lehmann R, Schleicher ED: High glucose-induced transforming growth factor-β1 production is mediated by the hexosamine pathway in porcine glomerular mesangial cells. J Clin Invest 101: 160–169, 1998
    OpenUrlCrossRefPubMed
  135. ↵
    Shankland SJ, Scholey JW, Ly H, Thai K: Expression of transforming growth factor-β1 during diabetic renal hypertrophy. Kidney Int 46: 430–442, 1994
    OpenUrlCrossRefPubMed
  136. ↵
    Sharma K, Ziyadeh FN: Renal hypertrophy is associated with upregulation of TGF-β1 gene expression in diabetic BB rat and NOD mouse. Am J Physiol 267: F1094–F1101, 1994
  137. ↵
    Pankewycz OG, Guan J-X, Bolton WK, Gomez A, Benedict JF: Renal TGF-β regulation in spontaneously diabetic NOD mice with correlations in mesangial cells Kidney Int 46: 748–758, 1994
    OpenUrlPubMed
  138. ↵
    Nakamura T, Fukui M, Ebihara I, Osada S, Nagaoka I, Tomino Y, Koide H: mRNA expression of growth factors in glomeruli from diabetic rats. Diabetes 42: 450–456, 1993
    OpenUrlAbstract/FREE Full Text
  139. ↵
    Hong SW, Isono M, Chen S, Iglesias-De La Cruz MC, Han DC, Ziyadeh FN: Increased glomerular and tubular expression of transforming growth factor-β1, its type II receptor, and activation of the Smad signaling pathway in the db/db mouse. Am J Pathol 158: 1653–1663, 2001
    OpenUrlCrossRefPubMed
  140. ↵
    Hill C, Flyvbjerg A, Gronbaek H, Petrik J, Hill DJ, Thomas CR, Sheppard MC, Logan A: The renal expression of transforming growth factor-beta isoforms and their receptors in acute and chronic experimental diabetes in rats. Endocrinology 141: 1196–1208, 2000
    OpenUrlCrossRefPubMed
  141. ↵
    Isono M, Mogyorosi A, Han DC, Hoffman BB, Ziyadeh FN: Stimulation of TGF-β type II receptor by high glucose in mouse mesangial cells and in diabetic kidney. Am J Physiol 278: F830–F838, 2000
    OpenUrl
  142. ↵
    Sharma K, Ziyadeh FN, Alzahabi B, McGowan TA, Kapoor S, Kurnik BRC, Kurnik PB, Weisberg LS: Increased renal production of transforming growth factor-β1 in patients with type II diabetes. Diabetes 26: 854–859, 1997
    OpenUrl
  143. ↵
    Iwano M, Kubo A, Atsushi K, Nishino T, Sato H, Nishioka H, Akai Y, Shiiki H, Kitamura K, Kanagawa K, Matsuo H, Dohi K: Quantification of glomerular TGF-β1 mRNA in patients with diabetes mellitus. Kidney Int 49: 1120–1126, 1996
    OpenUrlCrossRefPubMed
  144. ↵
    van Det NF, van den Born J, Tamsma JT, Verhagen NA, Berden JH, Bruijn JA, Daha MR, van der Woude FJ: Effects of high glucose on the production of heparan sulfate proteoglycan by mesangial and epithelial cells. Kidney Int 49: 1079–1089, 1996
    OpenUrlCrossRefPubMed
  145. ↵
    Chen S, Cohen MP, Lautenslager GT, Shearman CW, Ziyadeh FN: Glycated albumin stimulates TGF-β1 production and protein kinase C activity in glomerular endothelial cells. Kidney Int 59: 673–681, 2001
    OpenUrlCrossRefPubMed
  146. ↵
    Han DC, Isono M, Hoffman BB, Ziyadeh FN: High glucose stimulates proliferation and collagen type I synthesis in renal cortical fibroblasts: Mediation by autocrine activation of TGF-β. J Am Soc Nephrol 10: 1891–1899, 1999
    OpenUrlAbstract/FREE Full Text
  147. ↵
    Nakamura T, Miller D, Ruoslahti E, Border WA: Production of extracellular matrix by glomerular epithelial cells is regulated by transforming growth factor-β1. Kidney Int 41: 1213–1221, 1992
    OpenUrlCrossRefPubMed
  148. ↵
    Humes HD, Nakamura T, Cieslinski DA, Miller D, Emmons RV, Border WA: Role of proteoglycans and cytoskeleton in the effects of TGF-β1 on renal proximal tubule cells. Kidney Int 43: 575–584, 1993
    OpenUrlCrossRefPubMed
  149. ↵
    Roberts AB, McCune BK, Sporn MB: TGF-β: regulation of extracellular matrix. Kidney Int 41: 557–559, 1992
    OpenUrlCrossRefPubMed
  150. ↵
    Border WA, Noble NA: Transforming growth factor-β in tissue fibrosis. N Engl J Med 311: 1286–1292, 1994
  151. ↵
    Ziyadeh FN, Sharma K, Ericksen M, Wolf G: Stimulation of collagen gene expression and protein synthesis in murine mesangial cells by high glucose is mediated by autocrine activation of transforming growth factor-β. J Clin Invest 93: 536–542, 1994
  152. ↵
    Davies M, Thomas GJ, Martin J, Lovett DH: The purification and characterization of a glomerular-basement-membrane-degrading neutral proteinase from rat mesangial cells. Biochem J 251: 419–425, 1988
    OpenUrlAbstract/FREE Full Text
  153. ↵
    Marti HP, Lee L, Kashgarian M, Lovett DH: Transforming growth factor-β1 stimulates glomerular mesangial cell synthesis of the 72-kd type IV collagenase. Am J Pathol 144: 82–94, 1994
    OpenUrlPubMed
  154. ↵
    Eddy AA: Molecular insights into renal interstitial fibrosis. J Am Soc Nephrol 7: 2495–2508, 1996
    OpenUrlAbstract
  155. ↵
    Böttinger EP, Bitzer M: TGF-β Signaling in renal disease. J Am Soc Nephrol 13: 2600–2610, 2002
    OpenUrlAbstract/FREE Full Text
  156. ↵
    Isono M, Chen S, Hong SW, Iglesias-de la Cruz MC, Ziyadeh FN: Smad pathway is activated in the diabetic mouse kidney and Smad3 mediates TGF-β-induced fibronectin in mesangial cells. Biochem Biophys Res Commun 296: 1356–1365, 2002
    OpenUrlCrossRefPubMed
  157. ↵
    Mulder KM: Role of Ras and Mapks in TGFβ signaling. Cytokine Growth Factor Rev 11: 23–35, 2000
    OpenUrlCrossRefPubMed
  158. ↵
    Chin BY, Mohsenin A, Li SX, Choi AM, Choi ME: Stimulation of pro-α(1)(I) collagen by TGF-β1 in mesangial cells: Role pf the p38 MAPK pathway. Am J Physiol Renal Physiol 280: F495–F504, 2001
    OpenUrlPubMed
  159. ↵
    Wang L, Zhu Y, Sharma K: Transforming growth factor-β1 stimulates protein kinase A in mesangial cells. J Biol Chem 273: 8522–8527, 1998
    OpenUrlAbstract/FREE Full Text
  160. ↵
    Riser BL, Denichilo M, Cortes P, Baker C, Grondin JM, Yee J, Narins RG: Regulation of connective tissue growth factor activity in cultured rat mesangial cells and its expression in experimental diabetic glomerulosclerosis. J Am Soc Nephrol 11: 25–38, 2000
    OpenUrlAbstract/FREE Full Text
  161. ↵
    Twigg SM, Cao Z, McLennan SV, Burns WC, Brammar G, Forbes JM, Cooper ME: Renal connective tissue growth factor induction in experimental diabetes is prevented by aminoguanidine. Endocrinology 143: 4907–4915, 2002
    OpenUrlCrossRefPubMed
  162. ↵
    Ito Y, Aten J, Bende RJ, Oemar BS, Rabelink TJ, Weening JJ, Goldschmeding R: Expression of connective tissue growth factor in human renal fibrosis. Kidney Int 53: 853–861, 1998
    OpenUrlCrossRefPubMed
  163. ↵
    Adler SG, Kang SW, Feld S, Cha DR, Barba L, Striker L, Striker G, Riser BL, LaPage T, Nast CC: Glomerular mRNAs in human type 1 diabetes: biochemical evidence for microalbuminuria as a manifestation of diabetic nephropathy. Kidney Int 60: 2330–2336, 2001
    OpenUrlCrossRefPubMed
  164. ↵
    Park SK, Kim J, Seomun Y, Choi J, Kim DH, Han IO, Lee EH, Chung SK, Joo CK: Hydrogen peroxide is a novel inducer of connective tissue growth factor. Biochem Biophys Res Commun 284: 966–971, 2001
    OpenUrlCrossRefPubMed
  165. ↵
    Murphy M, McGinty A, Godson C: Protein kinases C: Potential targets for intervention in diabetic nephropathy. Curr Opin Nephrol Hypertens 7: 563–570, 1998
    OpenUrlCrossRefPubMed
  166. ↵
    Koya D, King G: Protein kinase C activation and the development of diabetic complications. Diabetes 47: 859–866, 1998
    OpenUrlAbstract
  167. ↵
    Igarashi A, Okochi H, Bradham DM, Grotendorst GR: Regulation of connective tissue growth factor gene expression in human skin fibroblasts and during wound repair. Mol Biol Cell 4: 637–645, 1993
    OpenUrlAbstract/FREE Full Text
  168. ↵
    Blom I, van Dijk A, Wieten L, Duran K, Ito Y, Kleij L, Denichilo M, Rabelink T, Weening J, Aten J, Goldschmeding R: In vitro evidence for differential involvement of CTGF, TGFbeta, and PDGF-BB in mesangial response to injury. Nephrol Dial Transplant 16: 1139–1148, 2001
    OpenUrlCrossRefPubMed
  169. ↵
    Ito Y, Goldschmeding R, Bende RJ, Claessen N, Chand MA, Kleij L, Rabelink TJ, Weening JJ, Aten J: Kinetics of connective tissue growth factor expression during experimental proliferative glomerulonephritis. J Am Soc Nephrol 12: 472–484, 2001
    OpenUrlAbstract/FREE Full Text
  170. ↵
    Chen Y, Blom IE, Sa S, Goldschmeding R, Abraham DJ, Leask A: CTGF expression in mesangial cells: Involvement of SMADs, MAP kinase, and PKC. Kidney Int 62: 1149–1159, 2002
    OpenUrlCrossRefPubMed
  171. ↵
    Grotendorst GR, Okochi H, Hayashi N: A novel transforming growth factor-β response element controls the expression of the connective tissue growth factor gene. Cell Growth Differ 7: 469–480, 1996
    OpenUrlAbstract
  172. ↵
    Holmes A, Abraham DJ, Sa S, Shiwen X, Black CM, Leask A: CTGF and SMADs, maintenance of scleroderma phenotype is independent of SMAD signaling. J Biol Chem 276: 10594–10601, 2001
    OpenUrlAbstract/FREE Full Text
  173. ↵
    Yokoi H, Mukoyama M, Sugawara A, Mori K, Nagae T, Makino H, Suganami T, Yahata K, Fujinaga Y, Tanaka I, Nakao K: Role of connective tissue growth factor in fibronectin expression and tubulointerstitial fibrosis. Am J Physiol Renal Physiol 282: F933–F942, 2002
    OpenUrlCrossRefPubMed
  174. ↵
    Grotendorst GR: Connective tissue growth factor: A mediator of TGF-β action on fibroblasts. Cytokine Growth Factor Rev 8: 171–179, 1997
    OpenUrlCrossRefPubMed
  175. ↵
    Duncan MR, Frazier KS, Abramson S, Williams S, Klapper H, Huang X, Grotendorst GR: Connective tissue growth factor mediates transforming growth factor beta-induced collagen synthesis: Down-regulation by cAMP. FASEB J 13: 1774–1786, 1999
    OpenUrlPubMed
  176. ↵
    Yokoi H, Sugawara A, Mukoyama M, Mori K, Makino H, Suganami T, Nagae T, Yahata K, Fujinaga Y, Tanaka I, Nakao K: Role of connective tissue growth factor in profibrotic action of transforming growth factor-β: A potential target for preventing renal fibrosis. Am J Kidney Dis 38 [Suppl]: S134–S138, 2001
    OpenUrlCrossRefPubMed
  177. ↵
    Gore-Hyer E, Shegogue D, Markiewicz M, Lo S, Hazen-Martin D, Greene EL, Grotendorst G, Trojanowska M: TGF-β and CTGF have overlapping and distinct fibrogenic effects on human renal cells. Am J Physiol Renal Physiol 283: F707–F716, 2002
    OpenUrlCrossRefPubMed
  178. ↵
    Wang S, Denichilo M, Brubaker C, Hirschberg R: Connective tissue growth factor in tubulointerstitial injury of diabetic nephropathy. Kidney Int 60: 96–105, 2001
    OpenUrlCrossRefPubMed
  179. ↵
    Crean JKG, Finlay D, Murphy M, Moss C, Godson C, Martin F, Brady HR: The role of p42/44 MAPK and protein kinase B in connective tissue growth factor induced extracellular matrix protein production, cell migration, and actin cytoskeletal rearrangement in human mesangial cells. J Biol Chem 277: 44187–44194, 2002
    OpenUrlAbstract/FREE Full Text
  180. ↵
    Throckmorton DC, Brogden AP, Min B, Rosmussen H, Kashgarian M: PDGF and TGF-β mediate collagen production by mesangial cells exposed to advanced glycosylation end products. Kidney Int 48: 111–117, 1995
    OpenUrlCrossRefPubMed
  181. ↵
    Di Paolo S, Gesualdo L, Ranieri E, Grandaliano G, Schena FP: High glucose concentration induces the overexpression of transforming growth factor-β through the activation of a platelet-derived growth factor loop in human mesangial cells. Am J Pathol 149: 2095–2106, 1996
    OpenUrlPubMed
  182. ↵
    Doi T, Vlassara H, Kirstein M, Yamada Y, Striker GE, Striker LJ: 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: 2873–2877, 1992
    OpenUrlAbstract/FREE Full Text
  183. ↵
    Flyvbjerg A: Potential use of growth hormone receptor antagonist in the treatment of diabetic kidney disease. Growth Horm IGF Res 11 [Suppl A]: S1150119, 2001
    OpenUrl
  184. ↵
    Chen NY, Chen WY, Bellush L, Yang CW, Striker LJ, Striker GE, Kopchick JJ: Effects of streptozotocin treatment in growth hormone (GH) and GH antagonist transgenic mice. Endocrinology 136: 660–667, 1995
    OpenUrlCrossRefPubMed
  185. ↵
    Esposito C, Liu ZH, Striker GE, Phillips C, Chen NY, Chen WY, Kopchick JJ, Striker LJ: Inhibition of diabetic nephropathy by a GH antagonist: A molecular analysis. Kidney Int 50: 506–514, 1996
    OpenUrlCrossRefPubMed
  186. ↵
    Bellush LL, Doublier S, Holland AN, Striker LJ, Striker GE, Kopchick JJ: Protection against diabetes-induced nephropathy in growth hormone receptor/binding protein gene-disrupted mice. Endocrinology 141: 163–168, 2000
    OpenUrlCrossRefPubMed
  187. ↵
    Doi T, Striker LJ, Quaife C, Conti FG, Palmiter R, Behringer R, Brinster R, Striker GE: Progressive glomerulosclerosis develops in transgenic mice chronically expressing growth hormone and growth hormone releasing factor but not in those expressing insulinlike growth factor-1. Am J Pathol 131: 398–403, 1988
    OpenUrlPubMed
  188. ↵
    Halldin MU, Tylleskar K, Hagenas L, Tuvemo T, Gustafsson J: Is growth hormone hypersecretion in diabetic adolescent girls also a daytime problem? Clin Endocrinol 48: 785–794, 1998
    OpenUrlCrossRefPubMed
  189. ↵
    Bereket A, Lang CH, Wilson TA: Alterations in the growth hormone-insulin-like growth factor axis in insulin dependent diabetes mellitus. Horm Metab Res 31: 172–181, 1999
    OpenUrlCrossRefPubMed
  190. ↵
    Doi SQ, Jacot TA, Sellitti DF, Hirszel P, Hirata MH, Striker GE, Striker LJ: Growth hormone increases inducible nitric oxide synthase expression in mesangial cells. J Am Soc Nephrol 11: 1419–1425, 2000
    OpenUrlAbstract/FREE Full Text
  191. ↵
    Trachtman H, Futterweit S, Pine E, Mann J, Valderrama E: Chronic diabetic nephropathy: Role of inducible nitric oxide synthase. Pediatr Nephrol 17: 20–29, 2002
    OpenUrlCrossRefPubMed
  192. ↵
    Piwien-Pilipuk G, Huo JS, Schwarz J: Growth hormone signal transduction. J Pediatr Endocrinol Metab 15: 771–786, 2002
    OpenUrlPubMed
  193. ↵
    Pugliese G, Pricci F, Romeo G, Pugliese F, Mene P, Giannini S, Cresci B, Galli G, Rotella CM, Vlassara H, Di Mario U: Upregulation of mesangial growth factor and extracellular matrix synthesis by advanced glycation end products via a receptor-mediated mechanism. Diabetes 46: 1881–1887, 1997
    OpenUrlAbstract/FREE Full Text
  194. ↵
    Moran A, Brown DM, Kim Y, Klein DJ: Effects of IGF-I and glucose on protein and proteoglycan synthesis by human fetal mesangial cells in culture. Diabetes 40: 1346–1354, 1991
    OpenUrlAbstract/FREE Full Text
  195. ↵
    Schreiber BD, Hughes ML, Groggel GC: Insulin-like growth factor-1 stimulates production of mesangial cell matrix components. Clin Nephrol 43: 368–374, 1995
    OpenUrlPubMed
  196. ↵
    Gooch JL, Tang Y, Ricono JM, Abboud HE: Insulin-like growth factor-I induces renal cell hypertrophy via a calcineurin-dependent mechanism. J Biol Chem 276: 42492–42500, 2001
    OpenUrlAbstract/FREE Full Text
  197. ↵
    Flyvbjerg A, Frystyk J, Osterby R, Orskov H: Kidney IGF-I and renal hypertrophy in GH-deficient diabetic dwarf rats. Am J Physiol 262: E956–E962, 1992
  198. ↵
    Flyvbjerg A, Kessler U, Dorka B, Funk B, Orskov H, Kiess W: Transient increase in renal insulin-like growth factor binding proteins during initial kidney hypertrophy in experimental diabetes in rats. Diabetologia 35: 589–593, 1992
    OpenUrlCrossRefPubMed
  199. ↵
    Segev Y, Landau D, Marbach M, Schadeh N, Flyvbjerg A, Phillip M: 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, 1997
    OpenUrlAbstract
  200. ↵
    Flyvbjerg A, Bennett WF, Rasch R, Kopchick JJ, Scarlett JA: Inhibitory effect of a growth hormone receptor antagonist (G120K-PEG) on renal enlargement, glomerular hypertrophy, and urinary albumin excretion in experimental diabetes in mice. Diabetes 48: 377–382, 1999
    OpenUrlAbstract
  201. ↵
    Weiss O, Anner H, Nephesh I, Alayoff A, Burstyn M, Raz I: Insulin-like growth factor-I (IGF-I) and IGF-I receptor gene expression in the kidney of the chronically hypoinsulinemic rat and hyperinsulinemic rat. Metabolism 44: 982–986, 1995
    OpenUrlCrossRefPubMed
  202. ↵
    Pillion DJ, Haskell JF, Meezan E: Distinct receptors for insulin-like growth factor I in rat renal glomeruli and tubules. Am J Physiol 255: E504–E512, 1988
  203. ↵
    Werner H, Shen-Orr Z, Stannard B, Burguera B, Robertes CT Jr, LeRoith D: Experimental diabetes increases insulinlike growth factor I and II receptor concentration and gene expression in kidney. Diabetes 39: 1490–1497
  204. ↵
    Horney MJ, Shirley DW, Kurtz DT, Rosenzweig SA: Elevated glucose increases mesangial cell sensitivity to insulin-like growth factor I. Am J Physiol 274: F1045–1053, 1998
  205. ↵
    Balaram SK, Agarwal DK, Edwards JD: Insulin like growth factor-1 activates nuclear factor-κB and increases transcription of the intercellular adhesion molecule-1 gene in endothelial cells. Cardiovasc Surg 7: 91–97, 1999
    OpenUrlCrossRefPubMed
  206. ↵
    Lupia E, Elliot SJ, Lenz O, Zheng F, Hattori M, Striker GE, Striker LJ: IGF-1 decreases collagen degradation in diabetic NOD mesangial cells. Implications for diabetic nephropathy. Diabetes 48: 1638–1644, 1999
    OpenUrlAbstract
  207. ↵
    Ha H, Lee HB: Reactive oxygen species as glucose signaling molecules in mesangial cells cultured under high glucose. Kidney Int Suppl 77: S19–25, 2000
    OpenUrlPubMed
  208. ↵
    Catherwood MA, Powell LA, Anderson P, McMaster D, Sharpe PC, Trimble ER: Glucose-induced oxidative stress in mesangial cells. Kidney Int 61: 599–608, 2002
    OpenUrlCrossRefPubMed
  209. ↵
    Suzuki YJ, Forman HJ, Sevanian A: Oxidants as stimulators of signal transduction. Free Radic Biol Med 22: 269–285, 1997
    OpenUrlCrossRefPubMed
  210. ↵
    Lander HM, Tauras JM, Ogiste JS, Hori O, Moss RA, Schmidt AK: Activation of the receptor for advanced glycation end products triggers a p21(ras)-dependent mitogen-activated protein kinase pathway regulated by oxidant stress. J Biol Chem 272: 17810–17814, 1997
    OpenUrlAbstract/FREE Full Text
  211. ↵
    Simon AR, Rai U, Fanburg BL, Cochran BH: Activation of the JAK-STAT pathway by reactive oxygen species. Am J Physiol 275: C1640–1652, 1998
  212. ↵
    Scivittaro V, Ganz MB, Weiss MF: AGEs induce oxidative stress and activate protein kinase C-β (II) in neonatal mesangial cells. Am J Physiol Renal Physiol 278: F676–683, 2000
    OpenUrlPubMed
  213. ↵
    Ohba M, Shibanuma M, Kuroki T, Nose K: Production of hydrogen peroxide by transforming growth factor-β1 and its involvement in induction of egr-1 in mouse osteoblastic cells. J Cell Biol 126: 1079–1088, 1994
    OpenUrlAbstract/FREE Full Text
  214. ↵
    Palmer HJ, Paulson KE: Reactive oxygen species and antioxidants in signal transduction and gene expression. Nutr Rev 55: 353–361, 1997
    OpenUrlPubMed
  215. ↵
    Kislinger T, Fu C, Huber B, Qu W, Taguchi A, Du Yan S, Hofmann M, Yan SF, Pischetsrieder M, Stern D, Schmidt AM: N(epsilon)-(carboxymethyl)lysine adducts of proteins are ligands for receptor for advanced glycation end products that activate cell signaling pathways and modulate gene expression. J Biol Chem 274: 31740–31749
  216. ↵
    Nath KA, Grande J, Croatt A, Haugen J, Kim Y, Rosenberg ME: Redox regulation of renal DNA synthesis, transforming growth factor-β1 and collagen gene expression. Kidney Int 53: 367–381, 1998
    OpenUrlCrossRefPubMed
  217. ↵
    Iglesias-De La Cruz MC, Ruiz-Torres P, Alcami J, Diez-Marques L, Ortega-Velazquez R, Chen S, Rodriguez-Puyol M, Ziyadeh FN, Rodriguez-Puyol D: Hydrogen peroxide increases extracellular matrix mRNA through TGFβ in human mesangial cells. Kidney Int 59: 87–95, 2001
    OpenUrlCrossRefPubMed
  218. ↵
    Raugi GJ, Lovett DH: Thrombospondin secretion by cultured human glomerular mesangial cells. Am J Pathol 129: 364–372, 1987
    OpenUrlPubMed
  219. ↵
    Tada H, Tsukamoto M, Nakayama K, Isogai S: The changes of glomerular thrombospondin in STZ-diabetic rats and the effect of high glucose on the synthesis of it in cultured mesangial cells [Abstract]. Diabetes 41: 122, 1992
  220. ↵
    McGregor B, Colon S, Mutin M, Chignier E, Zech P, McGregor J: Thrombospondin in human glomerulosclerosis. A marker for inflammation and early fibrosis. Am J Pathol 144: 1281–1287, 1994
    OpenUrlPubMed
  221. ↵
    Mumby SM, Raugi GJ, Bornstein P: Interactions of thrombospondin with extracellular matrix proteins: Selective binding to type V collagen. J Cell Biol 98: 646–652, 1984
    OpenUrlAbstract/FREE Full Text
  222. ↵
    Lahav J, Lawler J, Gimbrone MA: Thrombospondin interactions with fibronectin and fibrinogen. Mutual inhibition in binding. Eur J Biochem 145: 151–156, 1984
    OpenUrlPubMed
  223. ↵
    Tada H, Kawai H, Ishii H, Nomura K, Urayama T, Isogai S: Thrombospondin modulates adhesion, proliferation and production of extracellular matrix in mesangial cells. Tohoku J Exp Med 177: 239–302, 1995
    OpenUrl
  224. ↵
    Murphy-Ullrich JE, Poczatek M: Activation of latent TGF-β by thrombospondin-I: Mechanisms and physiology. Cytokine Growth Factor Rev 11: 59–69, 2000
    OpenUrlCrossRefPubMed
  225. ↵
    Poczatek MH, Hugo C, Darley-Usmar V, Murphy-Ullrich JE: Glucose stimulation of transforming growth factor-β bioactivity in mesangial cells is mediated by thrombospondin-1. Am J Pathol 157: 1353–1363, 2000
    OpenUrlCrossRefPubMed
  226. ↵
    Yevdokimova N, Wahab NA, Mason RM: Thrombospondin-1 is the key activator of TGF-β1 in human mesangial cells exposed to high glucose. J Am Soc Nephrol 12: 703–712, 2001
    OpenUrlAbstract/FREE Full Text
  227. ↵
    Crawford SE, Stellmach V, Murphy-Ullrich JE, Ribeiro SM, Lawler J, Hynes RO, Boivin GP, Bouck N: Thrombospondin-1 is a major activator of TGF-β1 in vivo. Cell 93: 1159–1170, 1998
    OpenUrlCrossRefPubMed
  228. ↵
    Mogyorosi A, Ziyadeh FN: Increased decorin mRNA in diabetic mouse kidney and in mesangial and tubular cells cultured in high glucose. Am J Physiol 275: F827–F832, 1998
  229. ↵
    Vogel KG, Paulsson M, Heinegard D: Specific inhibition of type I and type II collagen fibrillogenesis by the small proteoglycan of tendon. Biochem J 223: 587–597, 1984
    OpenUrlAbstract/FREE Full Text
  230. ↵
    Border WA, Noble NA, Yamamoto T, Harper JR, Yamaguchi Y, Pierschbacher MD, Ruoslahti E: Natural inhibitor of transforming growth factor-beta protects against scarring in experimental kidney disease. Nature 360: 361–364, 1992
    OpenUrlCrossRefPubMed
  231. ↵
    Abdel Wahab N, Wicks SJ, Mason RM, Chantry A: Decorin suppresses transforming growth factor beta-induced expression of plasminogen activator inhibitor-1 in human mesangial cells through a mechanism that involves Ca2+-dependent phosphorylation of Smad2 at serine-240. Biochem J 362: 643–649, 2002
    OpenUrlCrossRefPubMed
  232. ↵
    Liu Z, Thompson K, Towle H: Carbohydrate regulation of the rat L-type pyruvate kinase gene requires two nuclear factors: LF-A1 and a member of the c-myc family. J Biol Chem 268: 12787–12795, 1993
    OpenUrlAbstract/FREE Full Text
  233. ↵
    Sharma K, McGowan TA: TGF-β in diabetic kidney disease: Role of novel signaling pathways. Cytokine Growth Factor Rev 11: 115–123, 2000
    OpenUrlCrossRefPubMed
  234. Liu C, Yao J, de Belle I, Huang R-P, Adamson E, Mercola D: The transcription factor EGR-1 suppresses transformation of human fibrosarcoma HT1080 cells by coordinated induction of transforming growth factor-β1, fibronectin and plasminogen activator inhibitor-1. J Biol Chem 274: 4400–4411, 1999
    OpenUrlAbstract/FREE Full Text
  235. Zhang S-L, Chen X, Filep JG, Tang S-S, Ingelfinger JR, Chan JSD: High glucose levels stimulate phosphorylation cAMP-response element binding protein, activating transcription factor-2 and angiotensinogen gene expression in renal proximal tubular cells [Abstract]. J Am Soc Nephrol 11: A2480, 2000
    OpenUrl
  236. Laherty CD, Gierman TM, Dixit VM: Characterization of the promoter region of the human thrombospondin gene. DNA sequences within the first intron increase transcription. J Biol Chem 264: 11222–11227, 1989
    OpenUrlAbstract/FREE Full Text
  237. Bogdan C: Nitric oxide and the regulation of gene expression. Trends Cell Biol 11: 66–75, 2001
    OpenUrlCrossRefPubMed
  238. Wang S, Shiva S, Poczatek MH, Darley-Usmar V, Murphy-Ullrich JE: Nitric oxide and cGMP-dependent protein kinase regulation of glucose-mediated thrombospondin 1-dependent transforming growth factor-β activation in mesangial cells. J Biol Chem 277: 9880–9888, 2002
    OpenUrlAbstract/FREE Full Text
  239. Kreisberg JI, Radnik RA, Kreisberg SH: Phosphorylation of cAMP responsive element binding protein after treatment of mesangial cells with high glucose plus TGFβ or PMA. Kidney Int 50: 805–810, 1996
    OpenUrlCrossRefPubMed
  240. Lee BH, Park SY, Kang KB, Park RW, Kim IS: NF-κB activates fibronectin gene expression in rat hepatocytes. Biochem Biophys Res Commun 297: 1218–1224, 2002
    OpenUrlCrossRefPubMed
  241. Virolle T, Monthouel MN, Djabari Z, Ortonne JP, Neneguzzi G, Aberdam D: Three activator protein-1-binding sites bound by the Fra-2. JunD complex cooperate for the regulation of murine laminin alpha3A (lama3A) promoter activity by transforming growth factor-β. J Biol Chem 273: 17318–17325, 1998
    OpenUrlAbstract/FREE Full Text
  242. Zhang W, Ou J, Inagaki Y, Greenwel P, Ramirez F: Synergistic cooperation between Sp1 and Smad3/Smad4 mediates transforming growth factor-β1 stimulation of α2(I)-collagen (COL1A2) transcription. J Biol Chem 275: 39237–39245, 2000
    OpenUrlAbstract/FREE Full Text
  243. Segawa M, Kayano K, Sakaguchi E, Okamoto M, Sakaida I, Okita K: Antioxidant, N-acetyl-L-cysteine inhibits the expression of the collagen α2(I) promoter in the activated human hepatic stellate cell line in the absence as well as the presence of transforming growth factor-β. Hepatol Res 24: 305–315, 2002
    OpenUrlCrossRefPubMed
  244. Takami H, Burbelo PD, Fukuda K, Chang HS, Phillips SL, Yamada Y: In: Extracellular Matrix in the Kidney, edited by Koide H, Hayashi T, Basel, Karger, 1994, 36–46
  245. Wahab NA, Parker S, Sraer JD, Mason RM: The decorin high glucose response element and mechanism of its activation in human mesangial cells. J Am Soc Nephrol 11: 1607–1619, 2000
    OpenUrlAbstract/FREE Full Text
  246. Yingling JM, Datto MB, Wong C, Frederik JP, Liberati N, Wang XF: Tumor suppressor Smad4 is a transforming growth factor beta-inducible DNA binding protein. Mol Cell Biol 17: 7019–7028, 1997
    OpenUrlAbstract/FREE Full Text
  247. Song CZ, Siok TE, Gelehrter TD: Smad4/DPC4 and Smad3 mediate transforming growth factor-beta (TGF-β) signaling through direct binding to a novel TGF-β-responsive element in the human plasminogen activator inhibitor-1 promoter. J Biol Chem 273: 29287–29290, 1998
    OpenUrlAbstract/FREE Full Text
  248. Botelho FM, Edwards DR, Richards CD: Oncostatin M stimulates c-Fos to bind a transcriptionally responsive AP-1 element within the tissue inhibitor of metalloproteinase-1 promoter. J Biol Chem 273: 5211–5218, 1998
    OpenUrlAbstract/FREE Full Text
  249. Verrecchia F, Chu ML, Mauviel A: Identification of novel TGF-beta/Smad gene targets in dermal fibroblasts using a combined cDNA microarray/promoter transactivation approach. J Biol Chem 275: 17058–17062, 2001
    OpenUrl
  250. ↵
    Hocevar BA, Brown TL, Howe PH: TGF-β induces fibronectin synthesis through a c-Jun N-terminal kinase-dependent, Smad4-indpendent pathway. EMBO J 18: 1345–1356, 1999
    OpenUrlCrossRef
  251. ↵
    Isono M, Chen S, Hong SW, Iglesias-de la Cruz MC, Ziyadeh FN: Smad pathway is activated in the diabetic mouse kidney and Smad3 mediates TGF-β-induced fibronectin in mesangial cells. Biochem Biophys Res Commun 296: 1356–1365, 2002
  252. ↵
    Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, Richardson MA, Topper JN, Gimbrone MA Jr, Wrana JL, Falb D: The MAD-related protein Smad7 associates with the TGFβ receptor and functions as an antagonist of TGFβ signaling. Cell 89: 1165–1173, 1998
  253. ↵
    Chen R, Huang C, Morinelli TA, Trojanowska M, Paul RV: Blockade of the effects of TGF-β1 on mesangial cells by overexpression of Smad7. J Am Soc Nephrol 13: 887–893, 2002
    OpenUrlAbstract/FREE Full Text
  254. ↵
    Wahab NA, Weston BS, Roberts T, Mason RM: Connective tissue growth factor and regulation of the mesangial cell cycle: Role in cellular hypertrophy. J Am Soc Nephrol 13: 2437–2445, 2002
    OpenUrlAbstract/FREE Full Text
  255. ↵
    Westermarck J, Kahari V-M: Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J 13: 781–792, 1999
    OpenUrlCrossRefPubMed
  256. ↵
    Kerr LD, Miller DB, Matrisian LM: TGF-β1 inhibition of transin/stromelysin gene expression is mediated through a Fos binding sequence. Cell 61: 267–278, 1990
    OpenUrlCrossRefPubMed
  257. ↵
    Gaire M, Magbanua Z, McDonnell S, McNeil L, Lovett DH: Structure and expression of the human gene for the matrix metalloproteinase matrilysin. J Biol Chem 269: 2032–2040, 1994
    OpenUrlAbstract/FREE Full Text
  258. ↵
    Marti HP, McNeil L, Thomas G, Davies M, Lovett D: Molecular characterization of a low-molecular-mass matrix metalloproteinase secreted by glomerular mesangial cells as PUMP-1. Biochem J 285: 899–905, 1992
  259. ↵
    Abdel Wahab N, Gibbs J, Mason RM: Regulation of gene expression by alternative polyadenylation and mRNA instability in hyperglycemic mesangial cells. Biochem J 336: 405–411, 1998
View Abstract
PreviousNext
Back to top

In this issue

Journal of the American Society of Nephrology: 14 (5)
Journal of the American Society of Nephrology
Vol. 14, Issue 5
1 May 2003
  • Table of Contents
  • Index by author
View Selected Citations (0)
Print
Download PDF
Sign up for Alerts
Email Article
Thank you for your help in sharing the high-quality science in JASN.
Enter multiple addresses on separate lines or separate them with commas.
Extracellular Matrix Metabolism in Diabetic Nephropathy
(Your Name) has sent you a message from American Society of Nephrology
(Your Name) thought you would like to see the American Society of Nephrology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Extracellular Matrix Metabolism in Diabetic Nephropathy
Roger M. Mason, Nadia Abdel Wahab
JASN May 2003, 14 (5) 1358-1373; DOI: 10.1097/01.ASN.0000065640.77499.D7

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Extracellular Matrix Metabolism in Diabetic Nephropathy
Roger M. Mason, Nadia Abdel Wahab
JASN May 2003, 14 (5) 1358-1373; DOI: 10.1097/01.ASN.0000065640.77499.D7
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Diabetic Nephropathy: The Magnitude of the Problem
    • Changes in Renal Extracellular Matrices in DN
    • Mesangial Cell Responses to Hyperglycemic Conditions In Vitro
    • Changes in Intermediary Metabolism in Response to Hyperglycemia
    • Attenuation of DN in Animal Models of Diabetes
    • Growth Factors and Hormones Affecting Matrix Protein Expression in DN
    • ROS and the Activation of Pathways Stimulating Matrix Accumulation
    • Other Factors Modulating the Activity of TGF-β
    • Hyperglycemia and the Regulation of Gene Expression
    • Concluding Comments
    • Acknowledgments
    • References
  • Figures & Data Supps
  • Info & Metrics
  • View PDF

More in this TOC Section

  • Polycystic Kidney Disease
  • Genotype–Phenotype Correlations in Autosomal Dominant and Autosomal Recessive Polycystic Kidney Disease
  • Role of Primary Cilia in the Pathogenesis of Polycystic Kidney Disease
Show more Frontiers in Nephrology

Cited By...

  • Myo-inositol Oxygenase (MIOX) Overexpression Drives the Progression of Renal Tubulointerstitial Injury in Diabetes
  • Whole-Genome Sequencing of Finnish Type 1 Diabetic Siblings Discordant for Kidney Disease Reveals DNA Variants associated with Diabetic Nephropathy
  • Transforming growth factor {beta} (TGF{beta}) and related molecules in chronic kidney disease (CKD)
  • A causal link between oxidative stress and inflammation in cardiovascular and renal complications of diabetes
  • NADPH Oxidase Nox5 Accelerates Renal Injury in Diabetic Nephropathy
  • High glucose induces platelet-derived growth factor-C via carbohydrate response element-binding protein in glomerular mesangial cells
  • myo-Inositol Oxygenase Overexpression Accentuates Generation of Reactive Oxygen Species and Exacerbates Cellular Injury following High Glucose Ambience: A NEW MECHANISM RELEVANT TO THE PATHOGENESIS OF DIABETIC NEPHROPATHY
  • High Glucose Up-regulates ADAM17 through HIF-1{alpha} in Mesangial Cells
  • Activation of Glycogen Synthase Kinase 3{beta} Ameliorates Diabetes-induced Kidney Injury
  • Effects of high glucose on integrin activity and fibronectin matrix assembly by mesangial cells
  • Genetic Analysis of Mesangial Matrix Expansion in Aging Mice and Identification of Far2 as a Candidate Gene
  • Genetic Deletion of Cell Division Autoantigen 1 Retards Diabetes-Associated Renal Injury
  • Inhibition of Src Kinase Blocks High Glucose-Induced EGFR Transactivation and Collagen Synthesis in Mesangial Cells and Prevents Diabetic Nephropathy in Mice
  • Transforming Growth Factor {beta} Integrates Smad 3 to Mechanistic Target of Rapamycin Complexes to Arrest Deptor Abundance for Glomerular Mesangial Cell Hypertrophy
  • Hyperglycemia Causes Renal Cell Damage via CCN2-Induced Activation of the TrkA Receptor: Implications for Diabetic Nephropathy
  • Knockout of Toll-Like Receptor-2 Attenuates Both the Proinflammatory State of Diabetes and Incipient Diabetic Nephropathy
  • Activation of Bone Morphogenetic Protein 4 Signaling Leads to Glomerulosclerosis That Mimics Diabetic Nephropathy
  • Endothelin-1 Increases Collagen Accumulation in Renal Mesangial Cells by Stimulating a Chemokine and Cytokine Autocrine Signaling Loop
  • The Protective Role of Smad7 in Diabetic Kidney Disease: Mechanism and Therapeutic Potential
  • Pathologic Classification of Diabetic Nephropathy
  • High ambient glucose induces angiotensin-independent AT-1 receptor activation, leading to increases in proliferation and extracellular matrix accumulation in MES-13 mesangial cells
  • Urinary Protein Profiles in a Rat Model for Diabetic Complications
  • Lack of Type VIII Collagen in Mice Ameliorates Diabetic Nephropathy
  • The Mesangial Cell Revisited: No Cell Is an Island
  • Serum and glucocorticoid regulated kinase and disturbed renal sodium transport in diabetes
  • IHG-1 Amplifies TGF-{beta}1 Signaling and Is Increased in Renal Fibrosis
  • RhoA/Rho-Kinase Contribute to the Pathogenesis of Diabetic Renal Disease
  • From Fibrosis to Sclerosis: Mechanisms of Glomerulosclerosis in Diabetic Nephropathy
  • AMP-activated Protein Kinase Inhibits Transforming Growth Factor-{beta}-induced Smad3-dependent Transcription and Myofibroblast Transdifferentiation
  • Oleate Induces a Myofibroblast-Like Phenotype in Mesangial Cells
  • Connective tissue growth factor (CTGF) promotes activated mesangial cell survival via up-regulation of mitogen-activated protein kinase phosphatase-1 (MKP-1)
  • Interstitial Vascular Rarefaction and Reduced VEGF-A Expression in Human Diabetic Nephropathy
  • TGF-beta1 Regulates the PINCH-1-Integrin-Linked Kinase-{alpha}-Parvin Complex in Glomerular Cells
  • Distinguishing diabetic nephropathy from other causes of glomerulosclerosis: an update
  • Endothelial Nitric Oxide Synthase Deficiency Produces Accelerated Nephropathy in Diabetic Mice
  • Mesangial Cell Hypertrophy by High Glucose Is Mediated by Downregulation of the Tumor Suppressor PTEN.
  • Mammalian Target of Rapamycin Pathway Blockade Slows Progression of Diabetic Kidney Disease in Rats
  • The Pathogenesis of Myocardial Fibrosis in the Setting of Diabetic Cardiomyopathy
  • Hepatocyte Growth Factor Is a Downstream Effector that Mediates the Antifibrotic Action of Peroxisome Proliferator-Activated Receptor-{gamma} Agonists
  • Modulation of renal-specific oxidoreductase/myo-inositol oxygenase by high-glucose ambience
  • Assessment of 115 Candidate Genes for Diabetic Nephropathy by Transmission/Disequilibrium Test
  • Mechanism of Perturbation of Integrin-Mediated Cell-Matrix Interactions by Reactive Carbonyl Compounds and Its Implication for Pathogenesis of Diabetic Nephropathy
  • Chronic Angiotensin II Receptor Blockade Reduces (Intra)Renal Vascular Resistance in Patients with Type 2 Diabetes
  • Connective Tissue Growth Factor Expressed in Tubular Epithelium Plays a Pivotal Role in Renal Fibrogenesis
  • Intravenous Administration of Hepatocyte Growth Factor Gene Ameliorates Diabetic Nephropathy in Mice
  • High Ambient Glucose Enhances Sensitivity to TGF-{beta}1 via Extracellular Signal--Regulated Kinase and Protein Kinase C{delta} Activities in Human Mesangial Cells
  • Hepatocyte Growth Factor Antagonizes the Profibrotic Action of TGF-{beta}1 in Mesangial Cells by Stabilizing Smad Transcriptional Corepressor TGIF
  • Endothelial Progenitor Cell Dysfunction: A Novel Concept in the Pathogenesis of Vascular Complications of Type 1 Diabetes
  • Google Scholar

Similar Articles

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Articles

  • Current Issue
  • Early Access
  • Subject Collections
  • Article Archive
  • ASN Annual Meeting Abstracts

Information for Authors

  • Submit a Manuscript
  • Author Resources
  • Editorial Fellowship Program
  • ASN Journal Policies
  • Reuse/Reprint Policy

About

  • JASN
  • ASN
  • ASN Journals
  • ASN Kidney News

Journal Information

  • About JASN
  • JASN Email Alerts
  • JASN Key Impact Information
  • JASN Podcasts
  • JASN RSS Feeds
  • Editorial Board

More Information

  • Advertise
  • ASN Podcasts
  • ASN Publications
  • Become an ASN Member
  • Feedback
  • Follow on Twitter
  • Password/Email Address Changes
  • Subscribe

© 2021 American Society of Nephrology

Print ISSN - 1046-6673 Online ISSN - 1533-3450

Powered by HighWire