Skip to main content

Main menu

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

User menu

  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

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

Advanced Search

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

Overview of Glucose Signaling in Mesangial Cells in Diabetic Nephropathy

Masakazu Haneda, Daisuke Koya, Motohide Isono and Ryuichi Kikkawa
JASN May 2003, 14 (5) 1374-1382; DOI: https://doi.org/10.1097/01.ASN.0000064500.89551.76
Masakazu Haneda
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Daisuke Koya
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Motohide Isono
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ryuichi Kikkawa
  • 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

Clinical studies such as the Diabetes Control and Complications Trial (DCCT) in subjects with type 1 diabetes (1), the UK Prospective Diabetes Study (UKPDS) in subjects with type 2 diabetes (2), and the Kumamoto Study in Japanese subjects with type 2 diabetes (3) clearly link longstanding hyperglycemia to diabetic nephropathy. Furthermore, the regression of glomerular pathologic changes by the maintenance of normoglycemia for 10 yr was shown in eight subjects with type 1 diabetes who were treated with pancreas transplantation (4). Therefore, the understanding of the hyperglycemia-related molecular pathogenesis of diabetic nephropathy is truly needed to provide further insight into therapeutic strategies for diabetic nephropathy.

Diabetic nephropathy is characterized histologically by an accumulation of extracellular matrix (ECM) proteins in the glomerular mesangium (5). Functionally, an increase in GFR, glomerular hyperfiltration, found in the early phase of diabetes has been proposed to be related to the future development of diabetic nephropathy (6,7⇓). These abnormalities could be caused by functional changes in diabetic glomeruli, particularly in glomerular mesangial cells because mesangial cells were found to be capable of producing ECM proteins (8,9⇓) and regulating GFR through their contractility (10,11⇓). An enhancement of the production of type IV collagen and fibronectin (12–14⇓⇓), a reduction of the contractile responsiveness to angiotensin II (15–17⇓⇓), and an overproduction of vasorelaxing eicosanoids (18–20⇓⇓) have been shown in diabetic glomeruli and mesangial cells cultured under high-glucose conditions. These functional changes in glomeruli and mesangial cells in diabetes are considered to be caused by the metabolic abnormalities in glomeruli and mesangial cells specific to diabetes. In this review, we would like to describe glucose-induced signaling abnormalities that lead to functional disturbances of mesangial cells in the diabetic milieu.

Glucose Transport into Mesangial Cells

The first step of glucose signaling is the transport of glucose into the cells through specific glucose transporters. Previous studies indicate that mesangial cells express only a small amount of insulin receptors or insulin-sensitive facilitative glucose transporters (GLUT-4). Alternatively, mesangial cells were found to express two types of glucose transporters, facilitative and sodium-coupled transporters (21), and brain type glucose transporter (GLUT-1) was shown to be a predominant isoform (22,23⇓). Therefore, excessive extracellular glucose in the diabetic milieu will easily enter the cells through GLUT-1 in an insulin-independent manner and induce various signaling pathways. The importance of GLUT-1 was suggested by the overexpression of GLUT-1 in mesangial cells, in which an excessive production of ECM proteins was observed even under normal glucose conditions (24,25⇓). Furthermore, glucose-induced fibronectin production was found to be reduced by antisense GLUT-1 in mesangial cells (26). We have found that transforming growth factor-β1 (TGF-β1), one of the cytokines playing an important role in the development of diabetic nephropathy, is able to stimulate glucose uptake in mesangial cells by inducing the expression of mRNA and protein of GLUT1 (27). Our results also indicate that endogenous TGF-β1 produced by mesangial cells under high-glucose conditions stimulates glucose uptake and thus may accelerate glucose-induced metabolic abnormalities in mesangial cells.

Glucose-Induced Signaling Pathways: Metabolic Pathways

Glucose that enters the cells will be metabolized mainly by the glycolytic pathway. However, in the presence of excessive glucose, it will also be metabolized by various pathways and activate various signaling pathways indicated in Figure 1. An increase in the entry of glucose into the polyol pathway, the diacylglycerol (DAG) synthetic pathway, and the hexosamine pathway was found in mesangial cells cultured under high-glucose conditions.

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

Figure 1. Glucose-induced signaling pathways. Glucose is transported to the cells through GLUT-1 and metabolized mainly by glycolytic pathway. Increased de novo synthesis of diacylglycerol (DAG) results in the activation of protein kinase C (PKC), mitogen-activated kinases (MAPK). Glucose is also converted to sorbitol by aldose reductase (AR) and then to fructose by sorbitol dehydrokinase (SDH). The hexosamine biosynthetic pathway merges from glycolysis using fructose-6-phosphate (F-6-P) to form glucosamine-6-phosphate (GlcN-6-P) by the rate-limiting enzyme glutamine:fructose-6-phosphate-aminotransferase (GFAT). Activation of PKC-MAPK and uridine-5-diphosphate-N-acetylglucosamine (UDP-GlcNAc) is capable to stimulate certain genes, including transforming growth factor-β (TGF-β). Activation of TGF-β stimulates glucose uptake by inducing the expression of GLUT-1. Activation of these signaling pathways induces the expression of extracellular matrix (ECM) proteins leading to the expansion of glomerular mesangium. G-6-P, glucose-6-phosphate; F-1,6–2P, fructose-1, 6-biphosphate; DHAP, dihydroxyacetone phosphate; G-3-P, glycerol-3-phosphate; PA, phosphatidic acid; GAP, glyceraldehyde-3-phosphate; GlcNAc-6-P, N-acetylglucosamine-6-phosphate.

Polyol Pathway.

One of the important pathways is the polyol pathway. Glucose is converted to sorbitol by aldose reductase and then to fructose by sorbitol dehydrogenase. Since the Km value of aldose reductase for glucose is relatively high, the rate of sorbitol production appears to be dependent on intracellular glucose concentrations. Enzymes of the polyol pathway, aldose reductase and sorbitol dehydrogenase, have been identified in glomerular cells including mesangial cells (28), and the accumulation of sorbitol and concomitant depletion of myo-inositol was found in mesangial cells under high-glucose conditions (29). Several mechanisms by which the activation of polyol pathway causes functional derangement of the cells are proposed. One of them is an increase in the ratio of NADH/NAD+ resulting from the conversion of sorbitol to fructose. This increase in the ratio of NADH/NAD+ is proposed to be linked to de novo synthesis of diacylglycerol (DAG) resulting in the activation of protein kinase C (PKC), which will be described below. The role of the polyol pathway in the development of diabetic nephropathy has been examined using specific inhibitors of polyol pathway, aldose reductase inhibitors. Although aldose reductase inhibitors were shown to correct glomerular hyperfiltration and albuminuria in diabetic rats as well as in diabetic humans (30–33⇓⇓⇓) and the mesangial expansion in diabetic rats (34,35⇓), other reports have not been able to confirm the effect of aldose reductase inhibitor on albuminuria, hyperfiltration, or mesangial expansion in diabetic animals (36,37⇓). The study using transgenic mice overexpressing human aldose reductase revealed that only occlusion of renal vessels and deposits in Bowman’s capsule were seen in the kidney (38). However, another recent study in rats transgenic for aldose reductase showed that diabetes-induced albuminuria was completely prevented by the overexpression of aldose reductase, although the transgene was expressed in proximal tubular segments but not in glomeruli (39). Therefore, it remains unsolved whether the polyol pathway is in fact linked to the evolution of diabetic nephropathy.

PKC-MAPK Pathway.

PKC, a family of serine-threonine kinases, has at least eleven isoforms that can be categorized into classical PKC (α, β1, β2, γ), novel PKC (δ, ε, η, θ, μ), and atypical PKC (ζ, λ) on the basis of their common structural features (40).

The classical PKC enzymes contain two cysteine-rich zinc finger-like motifs (C1 region), which are essential for interaction with phorbol ester and diacylglycerol (DAG), and a Ca2+-binding domain (C2 region) in their regulatory domain. In the novel PKC enzymes, Ca2+ is not required because the C2 region is absent. Instead, they are activated by phosphatidylserine and DAG or phorbol esters. The atypical PKC enzymes, which lack the C2 region and one of the cysteine-rich zinc finger-like motifs in the C1 region, are not activated by Ca2+, DAG, or phorbol esters, but they are dependent on phosphatidylserine and activated by cis-unsaturated fatty acids.

PKC activation is involved in regulating a number of vascular functions such as vascular permeability (41), contractility (42), cell proliferation (43), extracellular matrix protein synthesis (44), and signal transduction cascade for hormones (45) and growth factors (46). We and others have reported that high glucose increases DAG levels and subsequently activates PKC in vascular cells and tissues (47–49⇓⇓). The mechanism responsible for the activation of PKC by high glucose appears to be related to an elevation of de novo DAG levels from the glycolytic intermediates, dihydroxyacetone phosphate and glyceraldehyde 3-phosphate (Figure 1). However, PKC activation by high glucose seems unlikely to be dependent on hydrolysis of phosphatidylinositol by phospholipase C and phosphatidylcholine by phospholipase D. This is supported by the data that inositol phosphate production, which is derived from hydrolysis of phosphatidylinositol, is unchanged in cultured aortic smooth muscle cells (50) and mesangial cells exposed to elevated glucose levels (51).

Other biochemical abnormalities by which high glucose and/or diabetes can produce diabetic nephropathy might be involved in PKC activation. The effect of the polyol pathway on high glucose-induced PKC activation was demonstrated by the fact that treatment with aldose reductase inhibitors prevented high glucose–induced PKC activation in mesangial cells (52,53⇓). Similarly, aldose reductase has been implicated to be an obligatory mediator upstream of PKC activation by high glucose in human lens epithelial cells (54). Binding of advanced-glycation endproducts (AGE) to receptor for AGE (RAGE) produces a cascade of cellular signaling, such as PKC activation. Indeed, AGE-induced PKC activation has been shown to stimulate collagen mRNA expression in human mesangial cells (55). Moreover, Amadori-modified glycated albumin can also stimulate PKC activation, followed by increased production of TGF-β protein and type IV collagen in glomerular endothelial cells, even in physiologic glucose concentrations (56). Recent data by Ha et al. (57) have demonstrated that inhibition of high glucose-induced PKC activation effectively abrogated reactive oxygen species generation and nuclear factor-κB (NF-κB) activity, resulting in decreasing monocyte chemoattractant protein-1 secretion in mesangial cells. Alternatively, activation of PKC could also affect AGE formation, possibly through reactive oxygen species generation (58). Taken together, there is accumulating evidence that the different biochemical abnormalities produced by high glucose in vitro and/or hyperglycemia in vivo can influence one another, because many of the glucose metabolites are important sources for the different metabolic pathways.

Among eleven PKC isoforms, PKC α, β1, β2, δ, and ε exhibit enhanced activation in the glomeruli of diabetic rats as well as mesangial cells exposed to high glucose by Western blot analysis and/or confocal microscopy (54,59,60⇓⇓). In addition, the activation of PKC-ζ isoforms has been shown in cultured mesangial cells exposed to high glucose (61), although the mechanism for activation of PKC-ζ, which is independent of DAG, is unclear. The differences in the findings obtained from many laboratories on the identity of the PKC isoform(s) activation by high glucose and/or diabetes might be due to differences in the methodology used to detect the presence/localization of the PKC isoforms as well as to measure their activity.

In the renal glomeruli, mesangial expansion and capillary basement membrane thickening, mainly composed of type IV collagen, fibronectin, and laminin, are classical manifestations of diabetic nephropathy. The effect of high glucose on the expression of fibronectin and type IV collagen can be prevented by general PKC inhibitors such as staurosporine or calphostin C (62,63⇓), and treatment with PKC agonists stimulates type IV collagen expression (63) and fibronectin accumulation (64), suggesting that the effects of high glucose on increasing production of extracellular matrix proteins are mediated through PKC activation. PKC activation increases the expression of TGF-β1, a prototypical multifunctional cytokine, which is one of the most important growth factors in the regulation of extracellular matrix protein accumulation in diabetic nephropathy. One possible mechanism by which high glucose increases the expression and synthesis of TGF-β1 and extracellular matrix proteins is by PKC’s actions on inducing transcription factors c-fos and c-jun, which form complexes for activated protein-1 (AP-1) binding site, because increased mRNA expression of c-fos and c-jun protooncogenes has been demonstrated in cultured mesangial cells exposed to high glucose (65) and in rat glomeruli after induction of diabetes (66). Moreover, high glucose–induced TGF-β promoter activation with the use of luciferase reporter was inhibited by a PKC inhibitor through decreasing AP-1 activity (67).

Mitogen-activated protein kinases (MAPK), including the extracellular signal-regulated protein kinase-1/2 (ERK 1/2), stress-activated c-Jun N-amino terminal kinase (JNK), and p38 MAPK, play a key role in the intracellular signal transduction cascade to integrate the transcription of genes responsible for a variety of cellular responses relevant to diabetic nephropathy such as cell growth, differentiation, and extracellular matrix synthesis (68). We and others have shown that ERK as well as p38 MAPK are activated in mesangial cells exposed to high glucose and in rat glomeruli of early type 1 diabetes (69–73⇓⇓⇓⇓). Recently, Whiteside et al. (74) have found that mesangial cell p38 MAPK activation in response to endothelin-1, angiotensin II, and platelet-derived growth factor is significantly augmented in high glucose media. In a type 2 diabetic model, ERK activity was reported to be significantly activated in renal cortex of db/db mice as compared with nondiabetic mice (75). The co-activation of PKC and MAPK by high glucose reinforces the notion that PKC plays an important role in the process of ERK activation. For example, we found that a general PKC inhibitor, calphostin C, was able to prevent the activation of ERK under high glucose (69). Furthermore, the specific inhibitor for PKC-β isoform LY 333531 can also inhibit hyperglycemia-induced glomerular ERK activation in diabetic rats (unpublished data by Kitada M, Koya D, Haneda M). The mechanism of high glucose-induced p38 MAPK in mesangial cells was recently shown to be mediated possibly via reactive oxygen species or upstream signaling proteins rather than changes in the amount of p38 MAPK and PKC activation (73,74⇓). In contrast, in vascular smooth muscle cells, PKC-δ dependent activation of p38 MAPK has been proposed (71).

One of the candidate targets of ERK is cytosolic phospholipase A2 (cPLA2), which was shown to be phosphorylated and activated by ERK (76). Indeed, the activities of cPLA2 were found to be enhanced in mesangial cells exposed to high glucose, and this activation was prevented by PD98059, an inhibitor of an upstream kinase of ERK (MEK) (69). Recent reports also suggest that the activation of ERK in mesangial cells under high-glucose conditions is responsible for the overproduction of TGF-β and ECM proteins such as type IV collagen and fibronectin, possibly through AP-1 activation (67,77⇓).

Moreover, ERK pathway was shown to be associated with mesangial cell hypertrophy through the induction of a newly identified protein, p8, which is induced in mesangial cells by endothelin as well as high glucose and in diabetic kidney (78). The functional role of p8, which is a helix-loop-helix protein with a nuclear localization signal with a modest sequence homology to high mobility group (HMG) protein, in mesangial cell hypertrophy was confirmed by employing p8 knock down using the recently developed method of RNA interference (78).

To understand the functional and pathophysiologic significance of PKC-MAPK activation, the effects of vitamin E (d-α tocopherol), thiazolidinediones, and a specific inhibitor for PKC-β isoform LY 333531 were tested on renal dysfunction and pathology in diabetic animals and in mesangial cells exposed to high glucose. Vitamin E and thiazolidinediones can prevent glomerular hyperfiltration as well as albuminuria in streptozotocin (STZ)-induced diabetic rats by inhibiting PKC-MAPK activation through decreasing DAG levels without changing plasma glucose levels (79). Oral administration of LY333531 to diabetic rats can also normalize GFR as well as albuminuria in parallel with inhibition of PKC activity without affecting DAG levels. Furthermore, LY333531 was able to prevent the diabetes-induced abnormalities in mRNA expression of TGF-β1, type IV collagen, and fibronectin in glomeruli of diabetic rats (79). In addition, an expansion of glomerular mesangium, one of the important histologic characters of diabetic nephropathy, was significantly ameliorated in db/db mice by the treatment with PKC-β inhibitor. The enhanced expression of TGF-β and ECM proteins in glomeruli of db/db mice was also shown to be prevented by the PKC-β inhibitor (80). As may be suspected, PKC-β inhibition with LY333531 treatment has been shown to attenuate the progression of diabetic nephropathy functionally and structurally in STZ-induced mRen-2 rats, which develop hypertension dependent on activation of the renin-angiotensin system, in addition to hyperglycemia (81). However, it should be noted that the activation of other PKC isoforms by high glucose and/or diabetes may also contribute to diabetic nephropathy. Further studies are needed to clarify the relevance of each PKC isoform, especially by the development of inhibitors specific for other PKC isoforms, in addition to PKC-β, and by other ways using knock-in and/or knock-out of each PKC isoform in mesangial cells or mice.

Because activation of PKC-MAPK pathway could play a crucial role in mediating the development and progression of diabetic nephropathy, the use of inhibitors of PKC and MAPK would be valuable therapeutic strategies. However, PKC-MAPK inhibition with nonspecific inhibitors may increase the risk of adverse effects because PKC-MAPK acts as a general signaling system in cellular function. Therefore, an inhibitor such as PKC-β inhibitor, which has specificity for a single cellular signaling, could potentially obviate most of the adverse effects. The ongoing phase 3 clinical trials with a PKC-β inhibitor may hopefully reveal in the near future if there are benefits in patients with diabetic vascular complications.

Hexoamine Pathway.

In the glycolytic pathway, glucose is converted to glucose-6-phosphate via hexokinase and subsequently to fructose-6-phosphate. Glucosamine-6-phosphate is then formed using glutamine as an amino donor under control of the rate-limiting enzyme glutamine:fructose-6-phosphate amidotransferase (GFAT). Glucosamine-6-phosphate is further converted to uridine-5-diphosphate-N-acetylglucosamine (UDP-GlcNAc), resulting in proteoglycan synthesis and O-linked glycoproteins (82).

Inhibition of GFAT using either azaserine or antisense oligonucleotides has been shown to abrogate high glucose–induced TGF-β1 overexpression and subsequent effects on mesangial cell proliferation and matrix production through decreasing glucosamine metabolites in mesangial cells (83). Interestingly, this pathway is also associated with PKC activation because exposure to glucosamine resulted in the translocation of PKC α, β, and ε to plasma membrane and GFAT inhibitor azaserine attenuated the high glucose-induced translocation of PKC-β (84). Recently, it was also reported that azaserine suppressed high glucose–induced PKC activation and plasminogen activator inhibitor-1 (PAI-1) expression in mesangial cells, which is correlated with increased extracellular matrix accumulation in various kidney disease including diabetic nephropathy (85). The glycosylated modulation of the transcription factor Sp1 by N-acetylglucosamine is proposed to explain the link between increased hexoamine pathway and high glucose-induced PAI-1 overexpression in vascular smooth muscle cells (86). In addition, protein kinase A (PKA) activity through high glucose-induced hexoamine pathway is also involved in extracellular matrix protein laminin synthesis in mesangial cells (87).

Glucose-Induced Signaling Pathways: The TGF-β System

Various cytokines and growth factors are considered to be upregulated as a consequence of metabolic pathways described above and to play an important role in the development and progression of diabetic nephropathy. Among them, TGF-β, which promotes renal cell hypertrophy and stimulates extracellular matrix accumulation, has been shown to mediate virtually all of the pathologic changes of diabetic kidney disease (88). Several studies have concluded that high glucose stimulates the production of TGF-β and other intermediary growth factors (89). Transcriptional activation of the TGF-β1 gene by high ambient glucose has been demonstrated in mouse mesangial cells (90). The promoter of the mouse or human TGF-β1 gene has a consensus nucleotide sequence termed “glucose response element,” CACGTG, that seems to be the target for transcriptional activation by high glucose (90). In addition, the TGF-β1 promoter also has multiple AP-1-like consensus sites that respond to phorbol-ester/PKC stimulation (91). As described above, increased activities of PKC and the MAP kinase cascades in high-glucose conditions are essential for regulation of the promoter activity of TGF-β1 through the activation of the AP-1 complex (67,77,91⇓⇓).

Studies employing neutralizing anti-TGF-β antibodies have provided convincing evidence that the prosclerotic and hypertrophic effects of high ambient glucose in cultured renal cells are largely mediated by autocrine production and activation of TGF-β. In particular, such studies have involved glomerular mesangial cells (92), glomerular epithelial cells (93), proximal tubular cells (94), and renal cortical fibroblasts (95). Additionally, high ambient glucose upregulates TGF-β receptor mRNAs and proteins in cultured mesangial cells and tubular epithelial cells (96,97⇓). Elevated renal TGF-β mRNA and protein levels have been found in various experimental animal models of diabetes mellitus such as STZ-diabetic rats or mice and db/db mice (98). This event coincides with the development of kidney hypertrophy, which is likely linked to the increased expression of TGF-β1 and type II TGF-β receptor (99) in the mouse kidney. Hyperglycemia is required for upregulation of TGF-β and extracellular matrix production because restoration of normoglycemia by insulin attenuates the renal cortical (99) and glomerular (100) expression of these molecules.

The Smad family of proteins has been recently identified as a predominant signal transducer of TGF-β (101). Binding of TGF-β to its type II receptor and subsequent recruitment of the type I receptor results in the phosphorylation and activation of receptor-regulated Smads (R-Smads), Smad2 and Smad3. After associating with a common-Smad (Co-Smad), Smad4, Smad complex translocates into the nucleus, where it regulates the expression of target genes through the binding to Smad-binding element (SBE) (102). In the kidney of diabetic db/db and STZ mice, both the nuclear localization of Smad3 and the nuclear binding to SBE as well as the expressions of TGF-β ligand and its receptor were shown to be increased (103,104⇓). Therefore, these observations indicated that the TGF-β system (consisting of TGF-β itself, its receptor, and the signaling pathway) was activated in the kidney of diabetes.

The importance of the renal TGF-β system in diabetic kidney disease is shown by the significant attenuation of glomerular and renal hypertrophy and the increase in TGF-β1, α1(IV) collagen, and fibronectin mRNAs achieved by short-term treatment of STZ-diabetic mice with neutralizing monoclonal antibodies of TGF-β (99). Long-term anti-TGF-β antibody treatment prevented the mesangial matrix expansion of diabetic glomerulosclerosis and, most importantly, preserved the creatinine clearance, showing that neutralization of TGF-β activity could prevent the progression of renal failure in diabetes (105). Furthermore, the administration of anti-TGF-β antibody to db/db mice with established diabetic nephropathy at 16 wk of age was able to reverse glomerular membrane thickening and mesangial matrix accumulation (106). Together, these studies highlight the importance of the renal TGF-β system in the manifestations of diabetic kidney disease such as renal hypertrophy, glomerulosclerosis, and renal insufficiency.

Recent studies performed in diabetic patients with various degrees of nephropathy also implicate the renal TGF-β system in the development of diabetic renal disease. All three isoforms of TGF-β were elevated in both the glomerular and tubulointerstitial compartments of patients with established diabetic nephropathy (107,108⇓). Furthermore, glomerular TGF-β1 mRNA was markedly increased in renal biopsy specimens from patients with proven diabetic kidney disease (109). Aortic, renal vein, and urinary levels of TGF-β were measured in type 2 diabetic patients undergoing elective coronary artery catheterization. The gradient of TGF-β1 concentration across the renal vascular bed was positive in the diabetic patients, indicating net renal production of TGF-β1 (110). These results support the conclusion that the kidneys of diabetic patients overproduce TGF-β1 protein. Interestingly, treatment with the angiotensin-converting enzyme inhibitor captopril lowered circulating TGF-β1 levels in patients with diabetic nephropathy who had previously enrolled in the Collaborative Study Group (111). Furthermore, the captopril-treated patients who had a decrease in the serum TGF-β1 level tended to have a better-preserved GFR over the ensuing 2-yr period. These results suggest that TGF-β1 plays an important role in the progression of diabetic nephropathy and that angiotensin-converting enzyme inhibitor therapy may protect the kidney by lowering TGF-β1 production.

In summary, recent studies described above identify various glucose-induced signaling pathways as candidates for future therapeutic strategies for diabetic nephropathy. When the rapid increase in the number of diabetic subjects with end-stage renal disease is considered, novel strategies applicable to human subjects need to be developed for early and effective interventions in the future.

  • © 2003 American Society of Nephrology

References

  1. ↵
    DCCT: The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetesmellitus. N Engl J Med 329: 977–986, 1993
    OpenUrlCrossRefPubMed
  2. ↵
    UKPDS: 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
  3. ↵
    Ohkubo Y, Kishikawa H, Araki E, Miyata T, Isami S, Motoyoshi S, Kojima Y, Furuyoshi N, Shichiri M: Intensive insulin therapy prevents the progression of diabetic microvascular complications in Japanese patients with non-insulin-dependent diabetes mellitus-a randomised prospective 6-year study. Diabetes Res Clin Pract 28: 103–117, 1995
    OpenUrlCrossRefPubMed
  4. ↵
    Fioretto P, Steffes MW, Sutherland DE, Goetz FC, Mauer M: Reversal of lesions of diabetic nephropathy after pancreas transplantation. N Engl J Med 339: 69–75, 1998
    OpenUrlCrossRefPubMed
  5. ↵
    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
  6. ↵
    Mogensen CE: Glomerular filtration rate and renal plasma flow in short term and long term juvenile diabetes mellitus. Scand J Clin Invest 28: 91–100, 1971
    OpenUrlCrossRefPubMed
  7. ↵
    Hostetter TH, Troy JL, Brenner BM: Glomerular hemodynamics in experimental diabetes mellitus. Kidney Int 19: 410–415, 1981
    OpenUrlCrossRefPubMed
  8. ↵
    Haralson MA: Collagen polymorphism in cultured rat kidney mesangial cells. Lab Invest 57: 513–523, 1987
    OpenUrlPubMed
  9. ↵
    Ardaillou N, Bellon G, Nivez M-P, Rakotoarison S, Ardaillou R: Quantification of collagen synthesis by cultured human glomerular cells. Biochem Biophys Res Comm 991: 445–452, 1989
    OpenUrl
  10. ↵
    Ausiello DA, Kreisberg JI, Roy C, Karnovsky MJ: Contraction of cultured rat glomerular cells of apparent mesangial origin after stimulation with angiotensin II and vasopression. J Clin Invest 65: 754–760, 1980
  11. ↵
    Blantz RC, Konnen KS, Tucker BJ: Angiotensin II effects upon the glomerular microcirculation and ultrafiltration coefficient of the rat. J Clin Invest 57: 419–434, 1976
  12. ↵
    Ayo SH, Radnik RA, Garoni JA, Glass IIWF, Kreisberg JI: High glucose causes an increase in extracellular matrix proteins in cultured mesangial cells. Am J Pathol 136: 1339–1348, 1990
    OpenUrlPubMed
  13. ↵
    Ayo SH, Radnik RA, Glass WFd, Garoni JA, Rampt ER, Appling DR, Kreisberg JI: Increased extracellular matrix synthesis and mRNA in mesangial cells grown in high-glucose medium. Am J Physiol 260: F185–F191, 1991
  14. ↵
    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
  15. ↵
    Kikkawa R, Kitamura E, Fujiwara Y, Arimura T, Haneda M, Y. S: Impaired contractile responsiveness of diabetic glomeruli to angiotensin II: A possible indication of mesangial dysfunction in diabetes mellitus. Biochem Biophys Res Commun 136: 1185–1190, 1986
    OpenUrlCrossRefPubMed
  16. ↵
    Mene P, Pugliese G, Pricci F, DiMario U, Cinotti GA, Pugliese F: High glucose inhibits cytosolic calcium signaling in cultured rat mesangial cells. Kidney Int 43: 585–591, 1993
    OpenUrlCrossRefPubMed
  17. ↵
    Haneda M, Kikkawa R, Koya D, Uzu T, Maeda S, Togawa M, Shigeta Y: Alteration of mesangial response to ANP and angiotensin II by glucose. Kidney Int 44: 518–526, 1993
    OpenUrlCrossRefPubMed
  18. ↵
    Schambelan M, Blake S, Sraer J, al e: Increased prostaglandin production by glomeruli isolated from rats with streptozotocin-induced diabetes mellitus. J Clin Invest 75: 404–412, 1985
  19. ↵
    Craven PA, Patterson MC, DeRubertis FR: Role of enhanced arachionate availability through phospholipase A2 pathway in mediation of increased prostaglandin synthesis by glomeruli from diabetic rats. Diabetes 37: 429–435, 1988
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Williams B, Schrier RW: Glucose-induced protein kinase C activity regulated arachidonic acid release and eicosanoid production by cultured glomerular mesangial cells. J Clin Invest 92: 2889–2896, 1993
  21. ↵
    Wakisaka M, He Q, Spiro MJ, Spiro RJ: Glucose entry into rat mesangial cells is mediated by both Na+-coupled and facilitative transporters. Diabetologia 38: 291–297, 1995
    OpenUrlCrossRefPubMed
  22. ↵
    Heilig CW, Liu Y, England RL, Freytag SO, Gilbert JD, Heilig KO, Zhu M, Concepcion LA, Brosius FCI: D-glucose stimulates mesangial cell GLUT1 expression and basal and IGF-1-sensitive glucose uptake in rat mesangial cells: Implications for diabetic nephropathy. Diabetes 46: 1030–1039, 1997
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Heilig CW, Concepcion LA, Riser BL, Riser BL, Freytag SO, Zhu M, Cortes P: Overexpression of glucose transporters in rat mesangial cells cultured in a normal glucose milieu mimics the diabetic phenotype. J Clin Invest 96: 1802–1814, 1995
  24. ↵
    Ewen ME, Sluss HK, Whitehouse LL, Livingstone DM: TGF-β inhibition of Cdk4 synthesis is linked to cell cycle arrest: Cell 74: 1009–1020, 1993
    OpenUrlCrossRefPubMed
  25. ↵
    Weigert C, Brodbeck K, Brocius IIIFC, Huber M, Lehmann R, Friess U, Facchin S, Aulwurm S, Haring HU, Schleicher ED, Heilig CW: Evidence for a novel TGF-b1-independent mechanism of fibronectin production in mesangial cells overexpressing glucose transporters. Diabetes 52: 527–535, 2003
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Heilig CW, Kreisberg JI, Freytag S, Murakami T, Ebina Y, Guo L, Heilig K, Loberg R, Qu X, Henry D, Brosius FCI: Antisense GLUT-1 protects mesangial cells from glucose induction of GLUT-1 and fibronectin expression. Am J Physiol 280: F657–F666, 2001
    OpenUrl
  27. ↵
    Inoki K, Haneda M, Maeda S, Koya D, Kikkawa R: TGF-b1 stimulates glucose uptake by enhancing GLUT1 expression in mesangial cells. Kidney Int 55: 1704–1712, 1999
    OpenUrlCrossRefPubMed
  28. ↵
    Kikkawa R, Umemura K, Haneda M, Kajiwara N, Maeda S, Nishimura C, Shigeta. Y: Identification and characterization of aldose reductase in cultured rat mesangial cells. Diabetes 41: 1165–1171, 1992
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Kikkawa R, Umemura K, Haneda M, Ari T, Ebata K, Shigeta Y: Evidence for existence of polyol pathway in cultured rat mesangial cells. Diabetes 40: 465–471, 1987
    OpenUrl
  30. ↵
    Bank N, Moywer P, Aynedjian HS, Wikes BM, Silverman S: Sorbinil prevents glomerular hyperperfusion in diabetic rats. Am J Physiol 256: F1000–F1006, 1989
  31. ↵
    Tilton RG, Chang K, Pugliese G, Eades DM, Province MA, Sherman WR, Kilo C, Williamson JR: Prevention of hemodynamic and vasucular albumin filtration changes in diabetic rats by aldose reductase inhibitors. Diabetes 37: 1258–1270, 1989
    OpenUrl
  32. ↵
    Goldfarb S, Ziyadeh FN, Kern EFO, Simmons DA: Effects of polyol-pathway inhibition and dietary myo-inositol on glomerular hemodynamic function in experimental diabetes mellitus in rats. Diabetes 40: 465–471, 1991
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Passariello N, Sepe J, Marrazzo G, Cicco AD, Peluso A, Pisano MCA, Sgambato S, Tesauro P, D’Onofrio F: Effect of aldose reductase inhibitor (tolrestat) on urinary albumin excretion rate and glomerular filtration rate in IDDM subjects with nephropathy. Diabetes Care 16: 789–795, 1993
    OpenUrlAbstract/FREE Full Text
  34. ↵
    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
  35. ↵
    Itagaki I, Shimizu K, Kamanaka Y, Ebata K, Kikkawa R, Haneda M, Shigeta Y: The effect of an aldose reductase inhibitor (Enalrestat) on diabetic nephropathy in rats. Diabetes Res Clin Pract 25: 147–154, 1994
    OpenUrlCrossRefPubMed
  36. ↵
    Daniels B, Hostetter T: Aldose reductase inhibition and glomerular abnormalities in diabetic rats. Diabetes 38: 981–986, 1989
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Korner A, Celsi G, Eklof A-C, Linne T, Persson B, Aperia A: Sorbinil does not prevent hyperfiltration, elevated ultrafiltration pressure and albuminuria in streptzotocin-diabetic rats. Diabetologia 35: 414–614, 1992
    OpenUrlCrossRefPubMed
  38. ↵
    Yamaoka T, Nishimura C, Yamashita K, Itakura M, Yamada T, Fujimoto J, Kokai Y: Acute onset of diabetic pathological changes in transgenic mice with human aldose reductase cDNA. Diabetologia 38: 255–261, 1995
    OpenUrlCrossRefPubMed
  39. ↵
    Ng DP, Hardy CL, Burns WC, Muggli EE, Kerr N, McCausland J, Alcorn D, Adams TE, Zajac JD, Larkins RG, Dunlop ME: Prevention of diabetes-induced albuminuria in transgenic rats overexpressing human aldose reductase. Endocrine 2002: 47–56, 2002
  40. ↵
    Nishizuka Y: Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science 258: 607–614, 1992
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Lynch JJ, Ferro TJ, Blumenstock FA, Brockenauer AM, Malik AM: Increased endothelial albumin permeability mediated by protein kinase C activation. J Clin Invest 85: 1991–1998, 1990
  42. ↵
    Rasmussen H, Forder J, Kojima I, Scriabine A: TPA-induced contraction of isolated rabbit vascular smooth muscle. Biochem Biophys Res Comm 122: 776–784, 1984
    OpenUrlCrossRefPubMed
  43. ↵
    Kariya K, Kawahara Y, Tsuda T, Fukuzaki H, Takai Y: Possible involvement of protein kinase C in platelet-derived growth factor-stimulated DNA synthesis in vasucular smooth muscle cells. Atherosclerosis 63: 251–255, 1987
    OpenUrlCrossRefPubMed
  44. ↵
    Huhtala P LTC, Tryggvason K: Structure of the human type IV collagen gene. J Bio Chem 265: 11077–11082, 1990
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Hachiya HL, Takayama S, White MF, King GL: Regulationof insulin receptor internalization in vascular endothelial cells by insulin and phorbol esters. J Biol Chem 262: 6417–6424, 1987
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Presta M, Marier JAM, Ragnotti G: The mitogenic signaling pathway but not plasminogen activator inducing pathway of basic fibroblast growth factor is mediated through protein kinase C in fetal bovine aortic endothelial cells. J Biol Chem 109: 1877–1884, 1989
    OpenUrl
  47. ↵
    Inoguchi T, Battan R, Handler E, Sportsman JR, Heath W, King GL: Preferential elevation of protein kinase C isoform βII and diacylglycerol levels in the aorta and heart of diabetic rats: Differential reversibility to glycemic control by islet cell transplantation. Proc Natl Acad Sci USA 89: 11059–11063, 1992
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Shiba T, Inoguchi T, Sportsman JR, Heath W, Bursell S, King GL: Correlation of diacylglycerol and protein kinase C activity in rat retina to retinal circulation. Am J Physiol 265: E783–E793, 1993
  49. ↵
    Craven PA, DeRubertis FR: Protein kinase C is activated in glomeruli from streptozotocin diabetic rats. J Clin Invest 83: 1667–1675, 1989
  50. ↵
    Xia P, Inoguchi T, Kern TS, Engerman RL, Oates PJ, King GL: Characterization of the mechanism for the chronic activation of diacylglycerol-protein kinase C pathway in diabetes and hypergalactosemia. Diabetes 43: 1122–1129, 1994
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Ayo SH, Radnik R, Garoni JA, Troyer DA, Kreisberg JI: High glucose increases diacylglycerol mass and activates protein kinase C in mesangial cell cultures. Am J Physiol 261: F571–F577, 1991
  52. ↵
    Kapor-Drezgic J, Zhou X, Babazono T, Dlugosz JA, Hohman T, Whiteside CI: Effect of high glucose on mesangial cell protein kinase C-delta and -epsilon is polyol pathway-dependent. J Am Soc Nephrol 10: 1193–1203, 1999
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Ramana KV, Friedrich B, Bhatnagar A, Srivastava SK: Aldose reductase mediates cytotoxic signals of hyperglycemia and TNF-alpha in human lens epithelial cells. FASEB J 17: 315–317, 2003
    OpenUrlCrossRefPubMed
  54. ↵
    Whiteside CI, Dlugosz JA: Mesamgial cell protein kinase C isozyme activation in the diabetic milieu. Am J Physiol 282: F975–F980, 2002
    OpenUrl
  55. ↵
    Kim YS, Kim BC, Song CY, Hong HK, Moon KC, Lee HS: Advanced glycation end products stimulate collagen mRNA synthesis in mesangial cells mediated by protein kinase C and transforming growth factor-beta. J Lab Clin Med 138: 59–68, 2001
    OpenUrlCrossRefPubMed
  56. ↵
    Chen S, Cohen MP, Lautenslager GT, Shearman CW, Ziyadeh FN: Glycated albumin stimulate TGF-beta 1 production and protein kinase C activity in glomerular endothelial cells. Kidney Int 59: 673–681, 2001
    OpenUrlCrossRefPubMed
  57. ↵
    Ha H, Yu MR, Choi YJ, Kitamura M, Lee HB: Role of high glucose-induced nuclear factor-kappa B activation in monocyte chemoattractant protein-1 expression by mesangial cells. J Am Soc Nephrol 13: 894–902, 2002
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Inoguchi T, Li P, Umeda F, Yu HY, Kakimoto M, Imamura M, Aoki T, Etoh T, Hashimoto T, Naruse M, Sano H, Utsumi H, Nawata H: High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C-dependent activation of NAD(P)H oxidase in cultured vascular cells. Diabetes 49: 1939–1945, 2000
    OpenUrlAbstract
  59. ↵
    Koya D, Jirousek MR, Lin YW, Ishii H, Kuboki K, King GL: Characterization of protein kinase C beta isoform activation on the gene expression of transforming growth factor-β, extracellular matrix components, and prostanoids in the glomeruli of diabetic rats. J Clin Invest 100: 115–126, 1997
    OpenUrlCrossRefPubMed
  60. ↵
    Babazono T, Kapor-Drezgic J, Dlugosz JA, Whiteside C: Altered expression and subcellular localization of diacylglycerol-sensitive protein kinase C isoforms in diabetic rat glomerular cells. Diabetes 47: 668–676, 1998
    OpenUrlAbstract
  61. ↵
    Kikkawa R, Haneda M, Uzu T, Koya D, Sugimoto T, Shigeta Y: Translocation of protein kinase C α and ζ in rat glomerular mesangial cells cultured under high glucose conditions. Diabetologia 37: 838–841, 1994
    OpenUrlCrossRefPubMed
  62. ↵
    Lin S, Sahai A, Chugh SC, Pan X, Wallner EI, Danesh FR, Lomasney JW, Kanwar YS: High glucose stimulates synthesis of ficronectin via a novel protein kinase C, Rap1b, and B-Raf signaling pathway. J Biol Chem 277: 41725–41735, 2002
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Fumo P, Kuncio GS, Ziyadeh FN: PKC and high glucose stimulate collagen alpha1(IV) transcriptional activity in a reporter mesangial cell line. Am J Physiol 267: F632–F638, 1994
  64. ↵
    Studer RK, Craven PA, DeRubertis FR: Role for protein kinase C in the mediation of increased fibronectin accumulation by mesangial cells grown in high glucose medium. Diabetes 42: 118–126, 1993
    OpenUrlAbstract/FREE Full Text
  65. ↵
    Kreisberg J, Radnik R, Ayo S, Garoni J, Saikumar P: High glucose elevates c-fos and c-jun transcripts and protein in mesangial cell culture. Kidney Int 46: 105–112, 1994
    OpenUrlCrossRefPubMed
  66. ↵
    Shankland SJ, Scholey JW: Expression of growth-related protooncogenes during diabetic renal hypertrophy. Kidney Int 47: 782–788, 1995
    OpenUrlCrossRefPubMed
  67. ↵
    Weigert C, Sauer U, Brodbeck K, Pfeiffer A, Haring HU, Schleicher ED: AP-1 protein mediate hyperglycemia-induced activation of the human TGF-β1 promoter in mesangial cells. J Am Soc Nephol 11: 2007–2016, 2000
    OpenUrlAbstract/FREE Full Text
  68. ↵
    Borkemeyer D, Sorokin A, Dunn MJ: Multiple intracellular MAP kinase signaling cascade. Kidney Int 49: 1187–1198, 1996
    OpenUrlCrossRefPubMed
  69. ↵
    Haneda M, Araki S, Togawa M, Sugimoto T, Isono M, Kikkawa R: Mitogen-activated protein kinase cascade is activated in glomeruli of diabetic rats and glomerular mesangial cells cultured under high glucose conditions. Diabetes 46: 847–853, 1997
    OpenUrlAbstract/FREE Full Text
  70. ↵
    Awazu M, Ishikura K, Hida M, Hoshiya M: Mechanisms of mitogen-activated protein kinase activation in experimental diabetes. J Am Soc Nephrol 10: 738–745, 1999
    OpenUrlAbstract/FREE Full Text
  71. ↵
    Igarashi M, Wakasaki H, Takahara N, Ishii H, Jiang ZY, Yamauchi T, Kuboki K, Meier M, Rhodes CJ, King GL: Glucose or diabetes activates p38 mitogen-activated protein kinase via different pathways. J Clin Invest 103: 185–195, 1999
    OpenUrlCrossRefPubMed
  72. ↵
    Kang SW, Adler SG, Lapage J, Natarajan R: p38 MAPK and MAPK kinase 3/6 mRNA and activities are increased in early diabetic glomeruli: Kidney Int 60: 543–552, 2001
    OpenUrlCrossRefPubMed
  73. ↵
    Wilmer WA, Dixon CL, Herbert C: Chronic exposure of human mesangial cells to high glucose environments activates the p38 MAPK pathway. Kidney Int 60: 858–871, 2001
    OpenUrlCrossRefPubMed
  74. ↵
    Tsiani E, Lekas P, Fantus IG, Dlugosz J, Whiteside C: High glucose-enhanced activation of mesangial cell p38 MAPK by ET-1, ANG II, and platelet-derived growth factor. Am J Phisiol Endocrinol Metab 282: E161–E169, 2002
    OpenUrlPubMed
  75. ↵
    Feliers D, Duraisamy S, Faulkner JL, Duch J, Lee AV, Abboud HE, Choundhury GG, Kasinath BS: Activation of renal signaling pathways in db/db mice with type 2 diabetes. Kidney Int 60: 495–504, 2001
    OpenUrlCrossRefPubMed
  76. ↵
    Lin L-L, Wartmann M, Lin AY, Knopf JL, Seth A, Davis RJ: cPLA2 is phosphorylated and activated by MAP kinase. Cell 72: 269–278, 1993
    OpenUrlCrossRefPubMed
  77. ↵
    Isono M, Iglesias-de la Cruz MC, Chen S, Hong SW, Ziyadeh FN: Extracellular signal-regulated kinase mediates stimulation of TGF-β1 and matrix by high glucose in mesangial cells. J Am Soc Nephrol 11: 2222–2230, 2000
    OpenUrlAbstract/FREE Full Text
  78. ↵
    Goruppi S, Bonventre JV, Kriakis M: Signaling pathways and late-onset gene induction associated with renal mesangial cell hypertrophy. EMBO J 21: 5427–5436, 2002
    OpenUrlAbstract
  79. ↵
    Koya D, Lee I-K, Ishii H, Kanoh H, King GL: Prevention of glomerular dysfunctions in diabetic rats by treatment of d-α-tocopherol. J Am Soc Nephrol 8: 426–435, 1997
    OpenUrlAbstract
  80. ↵
    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 beta inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J 14: 439–447, 2000
    OpenUrlCrossRefPubMed
  81. ↵
    Kelly DJ, Zhang Y, Hepper C, Gow RM, Jaworski K, Kemp BE, Wilkinson-Berka JL, Gilbert RE: Protein kinase C β inhibition attenuates the progression of experimental diabetic nephropathy in the presence of continued hypertension. Diabetes 52: 512–518, 2003
    OpenUrlAbstract/FREE Full Text
  82. ↵
    Schleicher ED, Weigert C: Role of hexosamine biosynthetic pathway in diabetic nephropathy. Kidney Int 58: [Suppl]: S13–S18, 2000
  83. ↵
    Kolm-Litty V, Sauer U, Nerlich A, Lehmann R, Schleicher ED: High glucose-induced transforming growth factor beta1 production is mediated by the hexosamine pathway in porcine glomerular mesangial cells. J Clin Invest 101: 160–169, 1998
    OpenUrlCrossRefPubMed
  84. ↵
    Kolm-Litty V, Tippmer S, Haring HU, Schleicher E: Glucosamine induces translocation of protein kinase C isozymes in mesangial cells. Exp Clin Endocrinol Diabetes 106: 377–383, 1998
    OpenUrlCrossRefPubMed
  85. ↵
    Goldberg HJ, Whiteside CI, Fantus IG: The hexosamine pathway regulates the plasminogen activator inhibitor-1 gene promoter and Sp1 transcriptional activation through protein kinase C β1 and δ. J Biol Chem 277: 33833–33841, 2002
    OpenUrlAbstract/FREE Full Text
  86. ↵
    Du XL, Edelstein D, Rossetti L, Fantus IG, Goldberg H, Ziyadeh FN, Wu J, Brownlee M: Hyperglycemia-induced mitochondrial superoxiside 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
  87. ↵
    Singh LP, Crook ED: Hexosamine regulation of glucose-mediated laminine synthesis in mesangial cells involves protein kinase A and C. Am J Physiol 279: F646–F654, 2000
    OpenUrl
  88. ↵
    Ziyadeh FN: Evidence for the involvement of transforming growth factor-β in the pathogenesis of diabetic kidney disease: Are Koch’s postulates fulfilled? Curr Pract Med 1: 87–89, 1998
  89. ↵
    Sharma K, Ziyadeh FN: Biochemical events and cytokine interactions linking glucose metabolism to the development of diabetic nephropathy. Semin Nephrol 17: 80–92, 1997
    OpenUrlPubMed
  90. ↵
    Hoffman BB, Sharma K, Zhu Y, Ziyadeh FN: Transcriptional activation of transforming growth factor-β1 in mesangial cell culture by high glucose concentration. Kidney Int 54: 1107–1116, 1998
    OpenUrlCrossRefPubMed
  91. ↵
    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
  92. ↵
    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
  93. ↵
    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
  94. ↵
    Rocco MV, Chen Y, Goldfarb S, Ziyadeh FN: Elevated glucose stimulates TGF-β gene expression and bioactivity in proximal tubule. Kidney Int 41: 107–114, 1992
    OpenUrlCrossRefPubMed
  95. ↵
    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
  96. ↵
    Isono M, Mogyorósi 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
  97. ↵
    Guh JY, Yang ML, Yang YL, Chang CC, Chuang LY: Captopril reverses high-glucose-induced growth effects on LLC-PK1 cells partly by decreasing transforming growth factor-beta receptor protein expressions. J Am Soc Nephrol 7: 1207–1215, 1996
    OpenUrlAbstract
  98. ↵
    Sharma K, Ziyadeh FN: Hyperglycemia and diabetic kidney disease. The case for transforming growth factor-β as a key mediator. Diabetes 44: 1139–1146, 1995
    OpenUrlAbstract/FREE Full Text
  99. ↵
    Sharma K, Jin Y, Guo J, Ziyadeh FN: Neutralization of TGF-β by anti-TGF-β antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in STZ-induced diabetic mice. Diabetes 45: 522–530, 1996
    OpenUrlAbstract/FREE Full Text
  100. ↵
    Park I-S, 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
  101. ↵
    Heldin C, Miyazono K, ten Dijke P: TGF-β signaling from cell membrane to nucleus through SMAD proteins. Nature 390: 465–471, 1997
    OpenUrlCrossRefPubMed
  102. ↵
    Massague J, Wotton D: Transcriptional control by TGF-β/Smad signaling. EMBO J 19: 1745–1754, 2000
    OpenUrlFREE Full Text
  103. ↵
    Hong SW, Isono M, Chen S, Iglesias-de la Cruz MC, Han DC, Ziyadeh FN: Increased glomerular and tubular expression of TGF-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
  104. ↵
    Isono M, Chen S, Hong SW, Iglesias-de la Cruz MC, Ziyadeh FN: Smad pathway is activated in diabetic mouse kidney and Smad3 mediates TGF-β-induced fibronectin in mesangial cells. Biochem Biophys Res Comm 296: 1356–1365, 2002
    OpenUrlCrossRefPubMed
  105. ↵
    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 expression by treatment with monoclonal anti-TGF-β antibody in db/db diabetic mice. Proc Natl Acad Sci USA 97: 8015–8020, 2000
    OpenUrlAbstract/FREE Full Text
  106. ↵
    Chen S, Iglesias-de la Cruz MC, Jim B, Hong SW, Isono M, Ziyadeh FN: Reversibility of established diabetic nephropathy by anti-TGF-β antibody in db/db mice. Biochem Biophys Res Comm 300: 16–22, 2003
    OpenUrlCrossRefPubMed
  107. ↵
    Yamamoto T, Nakamura T, Noble N, Ruoslahti E, Border W: Expression of transforming growth factor beta is elevated in human and experimental diabetic nephropathy. Proc Natl Acad Sci USA 90: 1814–1818, 1993
    OpenUrlAbstract/FREE Full Text
  108. ↵
    Yoshioka K, Takemura T, Murakami K, Okada M, Hino S, Miyamoto H, Maki S: Transforming growth factor-β protein and mRNA in glomeruli in normal and diseased human kidneys. Lab Invest 68: 154–163, 1993
    OpenUrlPubMed
  109. ↵
    Iwano M, Kubo A, Nishino T, Sato H, Nishioka H, Akai Y, Kurioka H, Fujii Y, Kanauchi M, Shiiki H, Dohi K: Quantification of glomerular TGF-β1 mRNA in patients with diabetes mellitus. Kidney Int 49: 1120–1126, 1996
    OpenUrlCrossRefPubMed
  110. ↵
    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 46: 854–859, 1997
    OpenUrlAbstract/FREE Full Text
  111. ↵
    Sharma K, Eltayeb BO, McGowan TA, Dunn SR, Alzahabi B, Rohde R, Ziyadeh FN, Lewis EJ: Captopril-induced reduction of serum levels of transforming growth factor-beta1 correlates with long-term renoprotection in insulin-dependent diabetic patients. Am J Kidney Dis 34: 818–823, 1999
    OpenUrlCrossRefPubMed
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.
Overview of Glucose Signaling in Mesangial Cells 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
Overview of Glucose Signaling in Mesangial Cells in Diabetic Nephropathy
Masakazu Haneda, Daisuke Koya, Motohide Isono, Ryuichi Kikkawa
JASN May 2003, 14 (5) 1374-1382; DOI: 10.1097/01.ASN.0000064500.89551.76

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Overview of Glucose Signaling in Mesangial Cells in Diabetic Nephropathy
Masakazu Haneda, Daisuke Koya, Motohide Isono, Ryuichi Kikkawa
JASN May 2003, 14 (5) 1374-1382; DOI: 10.1097/01.ASN.0000064500.89551.76
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Glucose Transport into Mesangial Cells
    • 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...

  • Proteasome subunit-{alpha} type-6 protein is post-transcriptionally repressed by the microRNA-4490 in diabetic nephropathy
  • High glucose induces platelet-derived growth factor-C via carbohydrate response element-binding protein in glomerular mesangial cells
  • Heparin Prevents Intracellular Hyaluronan Synthesis and Autophagy Responses in Hyperglycemic Dividing Mesangial Cells and Activates Synthesis of an Extensive Extracellular Monocyte-adhesive Hyaluronan Matrix after Completing Cell Division
  • Inhibition of Src Kinase Blocks High Glucose-Induced EGFR Transactivation and Collagen Synthesis in Mesangial Cells and Prevents Diabetic Nephropathy in Mice
  • Transcriptional and Post-translational Modulation of myo-Inositol Oxygenase by High Glucose and Related Pathobiological Stresses
  • Modification of Collagen IV by Glucose or Methylglyoxal Alters Distinct Mesangial Cell Functions
  • Lack of Type VIII Collagen in Mice Ameliorates Diabetic Nephropathy
  • Overexpression of Calmodulin in Pancreatic {beta} Cells Induces Diabetic Nephropathy
  • Tissue Gene Expression of Renin-Angiotensin System in Human Type 2 Diabetic Nephropathy
  • Nox4 NAD(P)H Oxidase Mediates Hypertrophy and Fibronectin Expression in the Diabetic Kidney
  • Role of Upstream Stimulatory Factors in Regulation of Renal Transforming Growth Factor-{beta}1
  • Hepatocyte Growth Factor Antagonizes the Profibrotic Action of TGF-{beta}1 in Mesangial Cells by Stabilizing Smad Transcriptional Corepressor TGIF
  • Regression of Advanced Diabetic Nephropathy by Hepatocyte Growth Factor Gene Therapy in Rats
  • 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 to ASN Journals

© 2022 American Society of Nephrology

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

Powered by HighWire