Kidney Res Clin Pract > Volume 32(1); 2013 > Article
Ghayur and Margetts: Transforming growth factor-beta and the glomerular filtration barrier

Abstract

The increasing burden of chronic kidney disease worldwide and recent advancements in the understanding of pathologic events leading to kidney injury have opened up new potential avenues for therapies to further diminish progression of kidney disease by targeting the glomerular filtration barrier and reducing proteinuria. The glomerular filtration barrier is affected by many different metabolic and immune-mediated injuries. Glomerular endothelial cells, the glomerular basement membrane, and podocytes—the three components of the filtration barrier—work together to prevent the loss of protein and at the same time allow passage of water and smaller molecules. Damage to any of the components of the filtration barrier can initiate proteinuria and renal fibrosis. Transforming growth factor-beta (TGF-β) is a pleiotropic cytokine strongly associated with the fibrogenic response. It has a known role in tubulointerstitial fibrosis. In this review we will highlight what is known about TGF-β and how it interacts with the components of glomerular filtration barrier and causes loss of function and proteinuria.

Keywords

Proteinuria, Podocyte, Glomerular endothelial cell, Transforming growth factor beta, Angiopoietin

Introduction

Proteinuria is a common finding in many kidney diseases such as diabetic nephropathy and chronic glomerulonephritis. Elevated urine protein is strongly associated with glomerulosclerosis, interstitial fibrosis, and progression of chronic kidney disease [1], and independently predicts worse clinical outcomes [2]. Most agents that reduce proteinuria have beneficial effects on long-term renal function [3]. Despite widespread use of drugs that decrease proteinuria, progression to end-stage kidney disease remains all too common.
Traditionally, our understanding of the pathogenesis of proteinuria focused on glomerular hypertension. Effective treatment has been aimed at the renin–angiotensin system, which reduces intraglomerular pressure [4]. Over the past decade, interest has shifted from this mechanical view to a cellular and molecular perspective. The glomerular filtration barrier consists of endothelial cells, a basement membrane, and a specialized epithelial cell—the podocyte (Fig. 1). Dysfunction in any component of the glomerular filtration barrier can lead to proteinuria. Increasingly, it has been recognized that there is a basal cross-talk between the endothelium and the podocytes, mainly involving angiogenic molecules such as vascular endothelial growth factor (VEGF) that maintain the health of the filtration barrier [5]. Perturbation of this signaling can lead to endothelial cell or podocyte injury. Podocyte injury manifests as retraction of the podocyte foot processes leading to effacement, loss of slit diaphragm proteins [6], dedifferentiation, detachment, and apoptosis [7]. Podocyte de-differentiation has been described as a process similar to epithelial to mesenchymal transition (EMT) and has been shown to occur in response to transforming growth factor-beta (TGF-β) [8].

Causes and consequences of proteinuria

Many common kidney diseases such as diabetes and chronic glomerulonephritis first manifest with structural glomerular injury and proteinuria [9] followed by tubulointerstitial fibrosis [10]. Proteinuria is a prognostic indicator in a variety of kidney diseases. In a large cohort study, Hemmelgarn et al. [2] demonstrated that the presence of even mild proteinuria (trace to 1+ on urine dipstick or 30–300 mg/g albumin to creatinine ratio) increased the risk of mortality and progressive renal failure independently of baseline renal function and other prognostic markers. Proteinuria is a common condition with significant implications with respect to population health. In the National Health and Nutrition Examination Survey study, 8.2% of the general US population have mild proteinuria, and 1.1% have overt proteinuria [11].
Although proteinuria is strongly linked epidemiologically to progressive chronic kidney disease, there is ongoing debate as to whether proteinuria is causally linked to tubulointerstitial fibrosis. When renal tubular cells are grown in culture and exposed to high levels of protein, the cells express inflammatory and fibrogenic cytokines [12]. These in vitro findings are supported by evidence from animal models where protein overload alone induces chronic renal injury [13]. Other hypothesized mechanisms to explain progressive interstitial fibrosis after glomerular injury include loss of peritubular capillaries and tubular hypoxia [14], [15], filtered cytokines [13], misdirected filtrate from sclerosed glomeruli [14], and interstitial inflammation [15].
Although there is some disagreement [16], the prevalent paradigm is that proteinuria is primarily a result of dysfunction of the glomerular filtration apparatus with a special focus on the podocyte. The podocyte is a terminally differentiated epithelial cell with a highly specialized structure that consists of foot processes that wrap around the glomerular capillaries. A series of transmembrane proteins link the foot processes, and comprise the ‘slit diaphragm’ structure that is crucial to proper function of this barrier [6].
If podocytes are injured, the expression of the slit diaphragm proteins decreases. For example, in diabetic nephropathy, the expression of nephrin decreases [17] and nephrin appears in the urine [18]. Later in the course of glomerular disease, podocytes detach from the basement membrane and also are found in the urine [19], [20]. Podocyte apoptosis is also a common finding in advanced glomerular injury [21].

Communication between components of the filtration barrier

It is increasingly clear that components of the glomerular filtration barrier interact with each other directly or through paracrine mediators (Fig. 2). For example, podocytes secrete vascular growth factors that maintain glomerular endothelial health [22]. Elevated levels of podocyte derived VEGF leads to a collapsing glomerulopathy, whereas decreased VEGF leads to endotheliosis (endothelial swelling and fibrin deposition) [22]. An anti-VEGF antibody used commonly in bowel and renal cancer has been shown to increase the risk for proteinuria and hypertension [23], suggesting a direct clinical effect of altering the podocyte/endothelial cell interaction.
Glomerular endothelial cells show evidence of dysfunction at an early stage of glomerular injury [24] and secrete TGF-β in response to VEGF stimulation [25]. Paracrine interaction between endothelium and podocytes involves other angiogenic factors such as angiopoietin- (ANGPT) 1 [26] and ANGPT-2 [27]. These are secreted by podocytes and impact on endothelial cells.
Although not directly part of the glomerular filtration apparatus, mesangial cells play an important role in paracrine signaling. Mesangial cells respond to high glucose [28] and pathogenic immunoglobulin A (IgA) [29] by secreting TGF-β. Khan et al. [30] have shown that mesangial cells can have a protective effect. TGF-β bound to the integrin αvβ8 is sequestered on mesangial cell membrane. When β8 integrin was knocked out, they observed higher levels of bioactive TGF-β, which resulted in endothelial cell apoptosis and proteinuria [30]. Mesangial cells also produce other paracrine signals such as stromal-derived factor-1, which affects podocyte function [31].
Integrins play a role in the interaction between podocytes and the glomerular basement membrane (GBM). Integrin α3β1 on podocytes allows adhesion with β2 laminin of the GBM. Interruption of this interaction leads to podocyte loss and glomerulosclerosis [32].

Role of angiopoietins in proteinuria

Angiopoietins are known to modify the effects of VEGF [33]. In quiescent vasculature, ANGPT1 is produced by perivascular cells and has a paracrine effect on endothelial cells. ANGPT1 binds to its receptor, tyrosine endothelial kinase (TEK), and signals through phosphatidylinositol-4,5 bisphosphate 3-kinase (PI3K)/Akt to maintain vascular integrity [34]. ANGPT2 similarly binds to the TEK receptor and is thought to be an antagonist for ANGPT1. In pathologic settings, VEGF and ANGPT2 are upregulated and lead to vascular destabilization, angiogenic sprouting, and new vessel formation [35]. In the quiescent glomerulus, ANGPT1 and VEGF are produced by podocytes and have protective and antiproteinuric effects on endothelial cells [26].
ANGPT2 has been associated with proteinuria and renal injury in several settings, including lupus nephritis [36] and diabetic nephropathy [4]. In previous work, Davis et al. [27] created a mouse model where ANGPT2 was overexpressed by a podocyte specific promoter. In this model, ANGPT2 overexpression led to proteinuria and decreased podocyte nephrin expression. The authors did not describe podocytes foot process effacement in response to ANGPT2, but they demonstrated endothelial cell dysfunction which was believed to underlie the proteinuric response.
In a recently described model of TGF-β overexpression in the glomerulus using an adenovirus [37], we found that ANGPT1 and ANGPT2 were both upregulated in the glomeruli from animals exposed to AdTGFB1, which is similar to observations by Campean et al. [38] in the Thy 1.1 model of glomerular injury. In unstimulated podocytes, we found no expression of TEK, but exposure to TGF-β increased TEK expression. We were able to block the effects of TGF-β on podocytes in culture with small interfering RNA directed against TEK, or with antibodies directed against TEK or ANGPT1. We hypothesized that TGF-β is required for podocyte dedifferentiation either as a direct cofactor or through the induction of TEK receptors on podocytes [37].

Transforming growth factor-beta

Much more is known about TGF-β than was known in late 1970 s when De Larco and Todaro [39] considered it a “factor” responsible for changing a cell's phenotype and called it sarcoma growth factor”. TGF-β has three isoforms in humans [40]. TGF-β is ubiquitously expressed and all cell types and all cells appear to respond to TGF-β [41]. In the glomerulus, TGF-β is produced by the podocytes and mesangial cells [42]. TGF-β is bound to a latency-associated peptide and sequestered in the extracellular matrix. Activation of TGF-β B is a complex and tightly regulated process [41], [43]. The mechanism of TGF-β activity at the cellular level is also complex, but the SMAD proteins are key signaling molecules for TGF-β [44].

TGF-β is involved in proteinuria and glomerulosclerosis

TGF-β is associated with fibrosis in many organ systems [45] including the kidney, where it can be induced by angiotensin [46], proteinuria [47], and hypoxia [48]. TGF-β induces elaboration of extracellular matrix components. Although controversial [49], TGF-β possibly induces EMT of renal tubular cells [50], [51]. Numerous studies have identified TGF-β as being upregulated during the course of progressive renal injury [52]. Similarly, inhibition of TGF-β has been shown to ameliorate renal injury [53].
TGF-β is not only involved in ECM accumulation, fibrosis, and progressive renal impairment, but also plays a role in changes to the glomerular filtration barrier and induction of proteinuria. There is substantial evidence to support this latter observation. Urinary TGF-β correlates with the degree of proteinuria [54]. Direct inhibition of TGF-β in models of renal disease reduces proteinuria [55], [56], [57].
Proteinuria was described in three animal models of TGF-β overexpression [58], [59], [60]. High levels of systemic TGF-β using a transgenic mouse model were shown to induce proteinuria, but the mechanism was not described [58]. Krag et al. [59] described a novel transgenic mouse with TGF-β driven from the Ren-1c promoter of the juxtaglomerular apparatus that developed albuminuria [59]. Wang et al. [60] used a hydrodynamic gene transfer model to overexpress TGF-β in the glomerulus and identified the role of Wnt/β-catenin in TGF-β-induced proteinuria.

TGF-β and podocyte dedifferentiation

The presence of the slit diaphragm as an important component of the glomerular filtration apparatus has been known for more than 30 years. Over the past 15 years, key molecular components of the slit diaphragm such as nephrin have been identified [61], the loss of which is associated with proteinuria [6]. Initially, this loss of epithelial markers was termed “de-differentiation” [62], but work by Sam et al. [63] later supported by Li et al. [8] identified that podocytes may undergo EMT in response to TGF-β.
Podocyte dedifferentiation is an early event in glomerular injury, whereas subsequent podocyte apoptosis and sloughing has been observed in advanced renal disease [7]. Notably, apoptosis appears to be a later finding that occurs after the onset of proteinuria [21]. Podocyte cell death is a complex process involving not only apoptosis, but also anoikis, autophagy, and, in some cases, necrosis [64].
Several pathways and growth factors are involved in this EMT process. As mentioned, TGF-β has been shown to induce podocyte EMT in vitro [8], [63], [65]. SNAIL is one of a family of zinc finger regulatory proteins shown to be an important mediator of EMT [66] and has been shown to suppress nephrin gene transcription in a model of puramycin-induced nephrosis [67]. Other pathways shown to be involved in podocyte EMT include mammalian target of rapamycin (mTOR) [68], [69], glycogen synthase kinase 3 β (GSK3B) [70], β-catenin [71], integrin-linked kinase (ILK) [72], and Notch [73]. PI3K has also been shown to be a key regulator of podocyte EMT, and PI3K inhibition was shown to block podocyte injury in an in vitro IgA model [74]. Recently, Finer et al. [75] have shown that podocyte injury induced by Adriamycin is mostly mediated by PI3K, and TGF-β is not involved in this process, but is involved in a delayed, fibrogenic response.
Notch is proving to be a key regulator of podocyte injury and apoptosis. Notch is a transmembrane receptor protein. On ligand binding, Notch undergoes a series of modifications including intracellular cleavage by gamma secretase [76]. The intracellular Notch fragment then acts as a transcriptional regulator. Notch interacts with the β-catenin and Wnt pathways during development of the kidney, but is quiescent in the adult kidney. During injury, TGF-β increases expression of the Notch ligand Jagged 1 and thus induces Notch signaling. In a seminal paper, Niranjan et al. [77] showed that overexpression of the intracellular Notch fragment in podocytes leads to severe proteinuria, glomerulosclerosis, and podocyte apoptosis. Using a gamma secretase inhibitor to block Notch signaling, renal injury could be reduced in a rodent model [77].
TGF-β mainly employs SMAD proteins to translate its signal; however, other signaling pathways have been shown to be activated by TGF-β. We have shown that in peritoneal mesothelial cells, EMT still occurs in mice lacking SMAD3, suggesting that non-SMAD pathways are critical for EMT [78]. This is supported by the observation of Wang et al. [79], who observed ongoing proteinuria and foot process effacement in SMAD3–/– mice rendered diabetic. The SMAD3–/– mice were protected from glomerulosclerosis and renal dysfunction.
EMT has been observed in urinary podocytes in patients with diabetes [80]. Although understanding podocyte EMT will provide valuable insights into the mechanisms of early glomerular injury progressive kidney disease, there are also therapeutic implications. For example, it has been demonstrated that TGF-β antagonists, specifically bone morphogenetic protein 7, can reverse EMT changes in renal tubular epithelium [50] and podocytes [81].

TGF-β and glomerular endothelial cells

Research into the biology of glomerular endothelial cells has perhaps been overshadowed by a strong interest in the podocyte. Early research suggested that the size of endothelial cells fenestrations is much larger than most plasma proteins (60–100 nm) and thus the endothelium should not form a significant filtration barrier. More recent work identified the importance of the glycocalyx, a dense negatively charged protein complex, as an important component of the glomerular filtration barrier [82]. Any injury that perturbs the integrity of this glycocalyx may lead to proteinuria [83].
Podocyte-derived VEGF has direct action on glomerular endothelial cells including proliferation and production of TGF-β [25]. VEGF also appears to regulate endothelial fenestrations [22]. Endothelial fenestrations were decreased in a biopsy study of Pima Indians with diabetes [19]. This decreased endothelial fenestration correlated with decreased glomerular filtration and increased albuminuria, whereas podocyte loss correlated with albumin excretion alone. There has been a direct association observed between podocyte injury and endothelial injury possibly mediated by loss of podocyte VEGF excretion [84].
Glomerular endothelial cells also respond to angiopoietins. Satchell et al. [26] have demonstrated that glomerular endothelial cells express the angiopoietin receptor TEK. In this study, ANGPT1 decreased protein passage through endothelial cell monolayers. As mentioned previously, overexpression of ANGPT2 in podocytes led to endothelial apoptosis and proteinuria [27]. El-Banawy et al. [85] measured ANGPT2 in patients with lupus and found that serum concentration correlated with proteinuria and disease severity.
A recently identified mechanism by which TGF-β induces endothelial cell dysfunction is endothelial to mesenchymal transition [86]. This is a similar biological process to EMT, and involves many of the same initiating factors and signaling pathways [86]. These transitioned endothelial cells are demonstrated to be the source of interstitial fibrosis in three well-known mouse models [87]. Although there is reasonable evidence that endothelial mesenchymal transition plays a role in interstitial fibrosis, few studies have evaluated glomerular endothelial cells. In one such study of streptozotocin-induced diabetic nephropathy, Li et al. [88] used an endothelial lineage traceable transgenic mouse to show that endothelial mesenchymal transition does occur in glomerular endothelial cells. However, this observation predated the onset of proteinuria in this model, so the authors thought that endothelial transition may play a role in glomerulosclerosis, but not proteinuria. This same study identified TGF-β as an in vitro mediator of endothelial mesenchymal transition [88].

TGF-β and the GBM

The GBM is a unique structure composed of laminin, type IV collagen, nidogen, and heparan sulfate proteoglycan. Both podocytes and endothelial cells have a role in development and maintenance of this structure [89]. Early changes in diabetic nephropathy include thickening of the GBM [90]. Krag et al. [91] have shown that mild hyperglycemia alone does not affect the GBM, but combined hyperglycemia and overexpression of TGF-β leads to pronounced GBM thickening. Wang et al. [79] found that the diabetes-induced GBM thickening was attenuated in mice deficient in the TGF-β signaling protein SMAD3.
The GBM is clearly an integral component of the glomerular filtration barrier; several genetic diseases that manifest as proteinuria have defects in proteins expressed in the GBM [89]. The structure and composition of the GBM can be altered by TGF-β, and this can potentially lead to further changes in the glomerular filtration barrier and proteinuria.

Therapeutic significance

The evidence implicating TGF-β B in the induction of proteinuria, glomerulosclerosis, and renal fibrosis is compelling. This makes TGF-β an important target in the prevention of progressive renal disease. Direct inhibition of TGF-β is a potential therapeutic strategy and a recent Phase 1 clinical trial of an anti-TGF-β antibody showed that a single infusion of this agent was well tolerated [92].
Many conditions that lead to end stage kidney disease are chronic processes that would necessitate long term inhibition of TGF-β. TGF-β is pleiotropic and has roles in wound healing, inflammation [93], and suppression of early cancers [94]. Long-term inhibition of TGF-β likely carries significant risks. Downstream pathways activated by TGF-β have become important therapeutic targets. For example, connective tissue growth factor is induced by TGF-β and is upregulated in patients with diabetic nephropathy [95]. In a Phase I clinical trial, the anti-connective tissue growth factor antibody FG-3019 was well tolerated and demonstrated a significant reduction in albuminuria in patients with diabetic nephropathy [96].
Another potential downstream target of TGF-β is the mammalian target of rapamycin (mTOR). We have demonstrated that mTOR is important in TGF-β -induced EMT, especially in the absence of a SMAD3 signal [78]. Although associated with increased risk of proteinuria in renal transplantation, recent studies have suggested that low-dose mTOR inhibition may have anti-proteinuric effects, at least in certain animal models [97].
TGF-β has been shown to activate the Wnt signaling pathway in podocytes [60]. Blockade of Wnt signaling in vivo blocked downstream signals involved in EMT such as SNAIL, and reduced TGF-β induced urinary protein excretion. Canonical Wnt signaling, through facilitation of β-catenin action, have been implicated in fibrosis of various organ systems [98] and treatment with a compound that specifically inhibits β-catenin (ICG-001) has been effective in animal models of lung and kidney fibrosis [99], [100].
Increasingly, both genetic and epigenetic processes have been identified in fibrogenesis [101]. Micro RNAs (miRNAs) are small, noncoding RNA fragments that modulate the expression of other coding mRNAs. TGF-β has been shown to increase the expression of miRNA 192 which then alters downstream signaling events. Blocking miRNA has recently been shown to have a beneficial effect in a mouse model of diabetic nephropathy [102]. Specifically, blocking miR-192 reduced albuminuria and renal fibrosis.

Summary

Proteinuria is a harbinger of progressive renal dysfunction in a wide range of common kidney diseases. Even low levels of proteinuria are associated with increased morbidity, mortality, and progression to end-stage renal disease [2]. Therapies, such as angiotensin pathway inhibitors, have been shown to reduce proteinuria and slow down the progressive decline in renal function. Despite this, further therapies are required. There is increasing evidence that TGF-β plays an important role in induction of proteinuria, along with subsequent glomerulosclerosis and renal interstitial fibrosis. Directly targeting TGF-β is an exciting approach to chronic renal disease. Further understanding of the TGF-β pathways involved will lead to a range of novel therapies that will reduce proteinuria and prevent progressive decline in renal function.

Conflicts of interest

No conflict of interest.

References

1. Ruggenenti P, Schieppati A, Remuzzi G. Progression, remission, regression of chronic renal diseases. Lancet 357:2001;1601–1608.
crossref pmid
2. Hemmelgarn BR, Manns BJ, Lloyd A, James MT, Klarenbach S, Quinn RR, Wiebe N, Tonelli M. Relation between kidney function, proteinuria, and adverse outcomes. JAMA 303:2010;423–429.
crossref pmid
3. Lattanzio MR, Weir MR. Does blockade of the renin-angiotensin-aldosterone system slow progression of all forms of kidney disease? Curr Hypertens Rep 12:2010;369–377.
crossref pmid
4. Gnudi L. Molecular mechanisms of proteinuria in diabetes. Biochem Soc Trans 36:2008;946–949.
crossref pmid
5. Ly J, Alexander M, Quaggin SE. A podocentric view of nephrology. Curr Opin Nephrol Hypertens 13:2004;299–305.
crossref pmid
6. Tryggvason K, Patrakka J, Wartiovaara J. Hereditary proteinuria syndromes and mechanisms of proteinuria. N Engl J Med 354:2006;1387–1401.
crossref pmid
7. Wolf G, Ziyadeh FN. Cellular and molecular mechanisms of proteinuria in diabetic nephropathy. Nephron Physiol 106:2007;26–31.
crossref
8. Li Y, Kang YS, Dai C, Kiss LP, Wen X, Liu Y. Epithelial-to-mesenchymal transition is a potential pathway leading to podocyte dysfunction and proteinuria. Am J Pathol 172:2008;299–308.
crossref pmid pmc
9. Remuzzi G, Ruggenenti P, Benigni A. Understanding the nature of renal disease progression. Kidney Int 51:1997;2–15.
crossref pmid
10. Abbate M, Zoja C, Rottoli D, Corna D, Tomasoni S, Remuzzi G. Proximal tubular cells promote fibrogenesis by TGF-beta1-mediated induction of peritubular myofibroblasts. Kidney Int 61:2002;2066–2077.
crossref pmid
11. Castro AF, Coresh J. CKD surveillance using laboratory data from the population-based National Health and Nutrition Examination Survey (NHANES). Am J Kidney Dis 53:2009;S46–S55.
crossref pmid pmc
12. Perico N, Benigni A, Remuzzi G. Nat Rev Drug Discov. Present and future drug treatments for chronic kidney diseases: evolving targets in renoprotection 7:2008;936–953.
13. Hirschberg R, Wang S. Proteinuria and growth factors in the development of tubulointerstitial injury and scarring in kidney disease. Curr Opin Nephrol Hypertens 14:2005;43–52.
crossref pmid
14. Kriz W, LeHir M. Pathways to nephron loss starting from glomerular diseases-insights from animal models. Kidney Int 67:2005;404–419.
crossref pmid
15. Abbate M, Zoja C, Remuzzi G. How does proteinuria cause progressive renal damage? J Am Soc Nephrol 17:2006;2974–2984.
crossref pmid
16. Comper WD, Hilliard LM, Nikolic-Paterson DJ, Russo LM. Disease-dependent mechanisms of albuminuria. Am J Physiol Renal Physiol 295:2008;F1589–F1600.
crossref pmid
17. Wang SX, Rastaldi MP, Patari A, Ahola H, Heikkila E, Holthofer H. Patterns of nephrin and a new proteinuria-associated protein expression in human renal diseases. Kidney Int 61:2002;141–147.
crossref pmid
18. Patari A, Forsblom C. Havana M, Taipale H, Groop PH, Holthofer H: Nephrinuria in diabetic nephropathy of type 1 diabetes. Diabetes 52:2003;2969–2974.
crossref pmid
19. Weil EJ, Lemley KV, Mason CC, Yee B, Jones LI, Blouch K, Lovato T, Richardson M, Myers BD, Nelson RG. Podocyte detachment and reduced glomerular capillary endothelial fenestration promote kidney disease in type 2 diabetic nephropathy. Kidney Int 82:2012;1010–1017.
crossref pmid pmc
20. Camici M. Urinary detection of podocyte injury. Biomed Pharmacother 61:2007;245–249.
crossref pmid
21. Menini S, Iacobini C, Oddi G, Ricci C, Simonelli P, Fallucca S, Grattarola M, Pugliese F, Pesce C, Pugliese G. Increased glomerular cell (podocyte) apoptosis in rats with streptozotocin-induced diabetes mellitus: role in the development of diabetic glomerular disease. Diabetologia 50:2007;2591–2599.
crossref pmid
22. Eremina V, Sood M, Haigh J, Nagy A, Lajoie G, Ferrara N, Gerber HP, Kikkawa Y, Miner JH, Quaggin SE. Glomerular-specific alterations of VEGF-A expression lead to distinct congenital and acquired renal diseases. J Clin Invest 111:2003;707–716.
crossref pmid pmc
23. Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med 358:2008;1129–1136.
crossref pmid pmc
24. Advani A, Gilbert RE. The endothelium in diabetic nephropathy. Semin Nephrol 32:2012;199–207.
crossref pmid
25. Li ZD, Bork JP, Krueger B, Patsenker E, Schulze-Krebs A, Hahn EG, Schuppan D. VEGF induces proliferation, migration, and TGF-beta1 expression in mouse glomerular endothelial cells via mitogen-activated protein kinase and phosphatidylinositol 3-kinase. Biochem Biophys Res Commun 334:2005;1049–1060.
crossref pmid
26. Satchell SC, Anderson KL, Mathieson PW. Angiopoietin 1 and vascular endothelial growth factor modulate human glomerular endothelial cell barrier properties. J Am Soc Nephrol 15:2004;566–574.
crossref pmid
27. Davis B, Dei Cas A, Long DA, White KE, Hayward A, Ku CH, Woolf AS, Bilous R, Viberti G, Gnudi L. Podocyte-specific expression of angiopoietin-2 causes proteinuria and apoptosis of glomerular endothelia. J Am Soc Nephrol 18:2007;2320–2329.
crossref pmid
28. Wu D, Peng F, Zhang B, Ingram AJ, Gao B, Krepinsky JC. Collagen I induction by high glucose levels is mediated by epidermal growth factor receptor and phosphoinositide 3-kinase/Akt signaling in mesangial cells. Diabetologia 50:2007;2008–2018.
crossref pmid
29. Lai KN, Leung JC, Chan LY, Saleem MA, Mathieson PW, Tam KY, Xiao J, Lai FM, Tang SC. Podocyte injury induced by mesangial-derived cytokines in IgA nephropathy. Nephrol Dial Transplant 24:2008;62–72.
crossref pmid
30. Khan S, Lakhe-Reddy S, McCarty JH, Sorenson CM, Sheibani N, Reichardt LF, Kim JH, Wang B, Sedor JR, Schelling JR. Mesangial cell integrin alphavbeta8 provides glomerular endothelial cell cytoprotection by sequestering TGF-beta and regulating PECAM-1. Am J Pathol 178:2011;609–620.
crossref pmid pmc
31. Ding M, Cui S, Li C, Jothy S, Haase V, Steer BM, Marsden PA, Pippin J, Shankland S, Rastaldi MP, Cohen CD, Kretzler M, Quaggin SE. Loss of the tumor suppressor Vhlh leads to upregulation of Cxcr4 and rapidly progressive glomerulonephritis in mice. Nat Med 12:2006;1081–1087.
crossref pmid
32. Chen CA, Hwang JC, Guh JY, Chang JM, Lai YH, Chen HC. Reduced podocyte expression of alpha3beta1 integrins and podocyte depletion in patients with primary focal segmental glomerulosclerosis and chronic PAN-treated rats. J Lab Clin Med 147:2006;74–82.
crossref pmid
33. Fiedler U, Augustin HG. Angiopoietins: a link between angiogenesis and inflammation. Trends Immunol 27:2006;552–558.
crossref pmid
34. Papapetropoulos A, Fulton D, Mahboubi K, Kalb RG, O'Connor DS, Li F, Altieri DC, Sessa WC. Angiopoietin-1 inhibits endothelial cell apoptosis via the Akt/survivin pathway. J Biol Chem 275:2000;9102–9105.
crossref pmid
35. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN, Yancopoulos GD. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 277:1997;55–60.
crossref pmid
36. Kumpers P, David S, Haubitz M, Hellpap J, Horn R, Brocker V, Schiffer M, Haller H, Witte T. The Tie2 receptor antagonist angiopoietin-2 facilitates vascular inflammation in systemic lupus erythematosus. Ann Rheum Dis 68:2008;1638–1643.
crossref pmid
37. Ghayur A, Liu L, Kolb M, Chawla A, Lambe S, Kapoor A, Margetts PJ. Adenovirus-mediated gene transfer of TGF-beta1 to the renal glomeruli leads to proteinuria. Am J Pathol 180:2012;940–951.
crossref pmid
38. Campean V, Karpe B, Haas C, Atalla A, Peters H, Rupprecht H, Liebner S, Acker T, Plate K, Amann K. Angiopoietin 1 and 2 gene and protein expression is differentially regulated in acute anti-Thy1.1 glomerulonephritis. Am J Physiol Renal Physiol 294:2008;F1174–F1184.
crossref pmid
39. De Larco JE, Todaro GJ. Growth factors from murine sarcoma virus-transformed cells. Proc Natl Acad Sci U S A 75:1978;4001–4005.
crossref pmid pmc
40. Ask K, Bonniaud P, Maass K, Eickelberg O, Margetts PJ, Warburton D, Groffen J, Gauldie J, Kolb M. Progressive pulmonary fibrosis is mediated by TGF-beta isoform 1 but not TGF-beta3. Int J Biochem Cell Biol 40:2008;484–495.
crossref pmid
41. Jenkins G. The role of proteases in transforming growth factor-beta activation. Int J Biochem Cell Biol 40:2008;1068–1078.
pmid
42. Pozzi A, Zent R. TGF-beta sequestration by mesangial cell integrin alphavbeta8: A novel mechanism of glomerular endothelial cell regulation. Am J Pathol 178:2011;485–489.
pmid pmc
43. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta activation. J Cell Sci 116:2003;217–224.
crossref pmid
44. Patel P, Sekiguchi Y, Oh KH, Patterson SE, Kolb MR, Margetts PJ. Smad3-dependent and -independent pathways are involved in peritoneal membrane injury. Kidney Int 77:2010;319–328.
crossref pmid
45. Gambaro G, Weigert C, Ceol M, Schleicher ED. Inhibition of transforming growth factor-beta 1 gene overexpression as a strategy to prevent fibrosis. [Review] [36 refs]. Contrib Nephrol 131:2001;107–113.
46. Wolf G. Renal injury due to renin-angiotensin-aldosterone system activation of the transforming growth factor-beta pathway. Kidney Int 70:2006;1914–1919.
crossref pmid
47. Eddy AA, Giachelli CM. Renal expression of genes that promote interstitial inflammation and fibrosis in rats with protein-overload proteinuria. Kidney Int 47:1995;1546–1557.
crossref pmid
48. Gunaratnam L, Morley M, Franovic A, de Paulsen N, Mekhail K, Parolin DA, Nakamura E, Lorimer IA, Lee S.. Hypoxia inducible factor activates the transforming growth factor-alpha/epidermal growth factor receptor growth stimulatory pathway in VHL(-/-) renal cell carcinoma cells. J Biol Chem 278:2003;44966–44974.
crossref pmid
49. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV, Valerius MT, McMahon AP, Duffield JS. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 176:2010;85–97.
crossref pmid pmc
50. Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz F, Kalluri R. BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med 9:2003;964–968.
crossref pmid
51. Nephrol Dial Transplant.
52. Bottinger EP. TGF-beta in renal injury and disease. Semin Nephrol 27:2007;309–320.
pmid
53. Border WA, Noble NA. TGF-beta in kidney fibrosis: a target for gene therapy. Kidney Int 51:1997;1388–1396.
pmid
54. Goumenos DS, Tsakas S, el Nahas AM, Alexandri S, Oldroyd S, Kalliakmani P, Vlachojannis JG. Transforming growth factor-beta(1) in the kidney and urine of patients with glomerular disease and proteinuria. Nephrol Dial Transplant 17:2002;2145–2152.
crossref pmid
55. Hou CC, Wang W, Huang XR, Fu P, Chen TH, Sheikh-Hamad D, Lan HY. Ultrasound-microbubble-mediated gene transfer of inducible Smad7 blocks transforming growth factor-beta signaling and fibrosis in rat remnant kidney. Am J Pathol 166:2005;761–771.
crossref pmid pmc
56. Ka SM, Huang XR, Lan HY, Tsai PY, Yang SM, Shui HA, Chen A. Smad7 gene therapy ameliorates an autoimmune crescentic glomerulonephritis in mice. J Am Soc Nephrol 18:2007;1777–1788.
crossref pmid
57. Saegusa Y, Sadakane C, Koseki J, Hasegawa Y, Shindo S, Maruyama H, Takeda S, Takeda H, Hattori T. TJN-331 improves anti-glomerular basement membrane nephritis by inhibiting the production of intraglomerular transforming growth factor-beta1. Biol Pharm Bull 33:2010;1349–1354.
crossref pmid
58. Kopp JB, Factor VM, Mozes M, Nagy P, Sanderson N, Böttinger EP, Klotman PE, Thorgeirsson SS. Transgenic mice with increased plasma levels of TGF-beta 1 develop progressive renal disease. Lab Invest 74:1996;991–1003.
pmid
59. Krag S, Osterby R, Chai Q, Nielsen CB, Hermans C, Wogensen L. TGF-beta1-induced glomerular disorder is associated with impaired concentrating ability mimicking primary glomerular disease with renal failure in man. Lab Invest 80:2000;1855–1868.
crossref pmid
60. Wang D, Dai C, Li Y, Liu Y. Canonical Wnt/beta-catenin signaling mediates transforming growth factor-beta1-driven podocyte injury and proteinuria. Kidney Int 80:2011;1159–1169.
crossref pmid pmc
61. Tryggvason K. Unraveling the mechanisms of glomerular ultrafiltration: nephrin, a key component of the slit diaphragm. J Am Soc Nephrol 10:1999;2440–2445.
pmid
62. Patek CE, Fleming S, Miles CG, Bellamy CO, Ladomery M, Spraggon L, Mullins J, Hastie ND, Hooper ML. Murine Denys-Drash syndrome: evidence of podocyte de-differentiation and systemic mediation of glomerulosclerosis. Hum Mol Genet 12:2003;2379–2394.
crossref pmid
63. Sam R, Wanna L, Gudehithlu KP, Garber SL, Dunea G, Arruda JA, Singh AK. Glomerular epithelial cells transform to myofibroblasts: early but not late removal of TGF-beta1 reverses transformation. Transl Res 148:2006;142–148.
crossref pmid
64. Tharaux PL, Huber TB. How many ways can a podocyte die? Semin Nephrol 32:2012;394–404.
crossref pmid
65. Herman-Edelstein M, Thomas MC, Thallas-Bonke V, Saleem M, Cooper ME, Kantharidis P. Dedifferentiation of immortalized human podocytes in response to transforming growth factor-beta: a model for diabetic podocytopathy. Diabetes 60:2011;1779–1788.
crossref pmid pmc
66. Nieto MA. The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3:2002;155–166.
crossref pmid
67. Matsui I, Ito T, Kurihara H, Imai E, Ogihara T, Hori M. Snail, a transcriptional regulator, represses nephrin expression in glomerular epithelial cells of nephrotic rats. Lab Invest 87:2007;273–283.
crossref pmid
68. Godel M, Hartleben B, Herbach N, Liu S, Zschiedrich S, Lu S, Debreczeni-Mór A, Lindenmeyer MT, Rastaldi MP, Hartleben G, Wiech T, Fornoni A, Nelson RG, Kretzler M, Wanke R, Pavenstädt H, Kerjaschki D, Cohen CD, Hall MN, Rüegg MA, Inoki K, Walz G, Huber TB. Role of mTOR in podocyte function and diabetic nephropathy in humans and mice. J Clin Invest 121:2011;2197–2209.
crossref pmid pmc
69. Inoki K, Mori H, Wang J, Suzuki T, Hong S, Yoshida S, Blattner SM, Ikenoue T, Rüegg MA, Hall MN, Kwiatkowski DJ, Rastaldi MP, Huber TB, Kretzler M, Holzman LB, Wiggins RC, Guan KL. mTORC1 activation in podocytes is a critical step in the development of diabetic nephropathy in mice. J Clin Invest 121:2011;2181–2196.
crossref pmid pmc
70. Boini KM, Amann K, Kempe D, Alessi DR, Lang F. Proteinuria in mice expressing PKB/SGK-resistant GSK3. Am J Physiol Renal Physiol 296:2009;F153–F159.
crossref pmid
71. Kato H, Gruenwald A, Suh JH, Miner JH, Barisoni-Thomas L, Taketo MM, Faul C, Millar SE, Holzman LB, Susztak K. Wnt/{beta}-catenin pathway in podocytes integrates cell adhesion, differentiation, and survival. J Biol Chem 286:2011;26003–26015.
crossref pmid pmc
72. Kang YS, Li Y, Dai C, Kiss LP, Wu C, Liu Y. Inhibition of integrin-linked kinase blocks podocyte epithelial-mesenchymal transition and ameliorates proteinuria. Kidney Int 78:2010;363–373.
crossref pmid pmc
73. Murea M, Park JK, Sharma S, Kato H, Gruenwald A, Niranjan T, Si H, Thomas DB, Pullman JM, Melamed ML, Susztak K. Expression of Notch pathway proteins correlates with albuminuria, glomerulosclerosis, and renal function. Kidney Int 78:2010;514–522.
crossref pmid pmc
74. Wang C, Liu X, Ke Z, Tang Y, Li CC, Li CM, Ye Z, Zhang J, Lou T. Mesangial medium from IgA nephropathy patients induces podocyte epithelial-to-mesenchymal transition through activation of the phosphatidyl inositol-3-kinase/Akt signaling pathway. Cell Physiol Biochem 29:2012;743–752.
crossref pmid
75. Finer G, Schnaper HW, Kanwar YS, Liang X, Lin HY, Hayashida T. Divergent roles of Smad3 and PI3-kinase in murine adriamycin nephropathy indicate distinct mechanisms of proteinuria and fibrogenesis. Kidney Int 82:2012;525–536.
crossref pmid pmc
76. Kato H, Susztak K. Repair problems in podocytes: Wnt, Notch, and glomerulosclerosis. Semin Nephrol 32:2012;350–356.
crossref pmid pmc
77. Niranjan T, Bielesz B, Gruenwald A, Ponda MP, Kopp JB, Thomas DB, Susztak K. The Notch pathway in podocytes plays a role in the development of glomerular disease. Nat Med 14:2008;290–298.
crossref pmid
78. Patel P, Sekiguchi Y, Oh KH, Patterson SE, Kolb MR, Margetts PJ. Smad3-dependent and -independent pathways are involved in peritoneal membrane injury. Kidney Int 77:2010;319–328.
crossref pmid
79. Wang A, Ziyadeh FN, Lee EY, Pyagay PE, Sung SH, Sheardown SA, Laping NJ, Chen S. Interference with TGF-beta signaling by Smad3-knockout in mice limits diabetic glomerulosclerosis without affecting albuminuria. Am J Physiol Renal Physiol 293:2007;F1657–F1665.
crossref pmid
80. Yamaguchi Y, Iwano M, Suzuki D, Nakatani K, Kimura K, Harada K, Kubo A, Akai Y, Toyoda M, Kanauchi M, Neilson EG, Saito Y. Epithelial-mesenchymal transition as a potential explanation for podocyte depletion in diabetic nephropathy. Am J Kidney Dis 54:2009;653–664.
crossref pmid
81. De Petris L, Hruska KA, Chiechio S, Liapis H. Bone morphogenetic protein-7 delays podocyte injury due to high glucose. Nephrol Dial Transplant 22:2007;3442–3450.
crossref pmid
82. Hjalmarsson C, Johansson BR, Haraldsson B. Electron microscopic evaluation of the endothelial surface layer of glomerular capillaries. Microvasc Res 67:2004;9–17.
crossref pmid
83. Nieuwdorp M, Mooij HL, Kroon J, Atasever B, Spaan JA, Ince C, Holleman F, Diamant M, Heine RJ, Hoekstra JB, Kastelein JJ, Stroes ES, Vink H. Endothelial glycocalyx damage coincides with microalbuminuria in type 1 diabetes. Diabetes 55:2006;1127–1132.
crossref pmid
84. Satchell SC. The glomerular endothelium emerges as a key player in diabetic nephropathy. Kidney Int 82:2012;949–951.
crossref pmid
85. El-Banawy HS, Gaber EW, Maharem DA, Matrawy KA. Angiopoietin-2, endothelial dysfunction and renal involvement in patients with systemic lupus erythematosus. J Nephrol 25:2012;541–550.
crossref pmid
86. van Meeteren LA, ten Dijke P. Regulation of endothelial cell plasticity by TGF-beta. Cell Tissue Res 347:2012;177–186.
crossref pmid
87. Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol 19:2008;2282–2287.
crossref pmid pmc
88. Li J, Qu X, Bertram JF. Endothelial-myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am J Pathol 175:2009;1380–1388.
crossref pmid pmc
89. Miner JH. Glomerular basement membrane composition and the filtration barrier. Pediatr Nephrol 26:2011;1413–1417.
crossref pmid pmc
90. Caramori ML, Kim Y, Huang C, Fish AJ, Rich SS, Miller ME, Russell G, Mauer M. Cellular basis of diabetic nephropathy: 1. Study design and renal structural-functional relationships in patients with long-standing type 1 diabetes. Diabetes 51:2002;506–513.
crossref pmid
91. Krag S, Nyengaard JR, Wogensen L. Combined effects of moderately elevated blood glucose and locally produced TGF-beta1 on glomerular morphology and renal collagen production. Nephrol Dial Transplant 22:2007;2485–2496.
crossref pmid
92. Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H, Rota S, Remuzzi G, Rump LC, Sellin LK, Heaton JP, Streisand JB, Hard ML, Ledbetter SR, Vincenti F. A phase 1, single-dose study of fresolimumab, an anti-TGF-beta antibody, in treatment-resistant primary focal segmental glomerulosclerosis. Kidney Int 79:2011;1236–1243.
crossref pmid pmc
93. Yang X, Letterio JJ, Lechleider RJ, Chen L, Hayman R, Gu H, Roberts AB, Deng C. Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF-beta. EMBO J 18:1999;1280–1291.
crossref pmid pmc
94. Connolly EC, Freimuth J, Akhurst RJ. Complexities of TGF-beta targeted cancer therapy. Int J Biol Sci 8:2012;964–978.
crossref pmid pmc
95.
96. Adler SG, Schwartz S, Williams ME, Arauz-Pacheco C, Bolton WK, Lee T, Li D, Neff TB, Urquilla PR, Sewell KL. Phase 1 study of anti-CTGF monoclonal antibody in patients with diabetes and microalbuminuria. Clin J Am Soc Nephrol 5:2010;1420–1428.
crossref pmid pmc
97. Kurdián M, Herrero-Fresneda I, Lloberas N, Gimenez-Bonafe P, Coria V, Grande MT, Boggia J, Malacrida L, Torras J, Arévalo MA, González-Martínez F, López-Novoa JM, Grinyó J, Noboa O. Delayed mTOR inhibition with low dose of everolimus reduces TGFbeta expression, attenuates proteinuria and renal damage in the renal mass reduction model. PLoS ONE 7:2012;e32516
crossref pmid pmc
98.
99. Hao S, He W, Li Y, Ding H, Hou Y, Nie J, Hou FF, Kahn M, Liu Y. Targeted inhibition of {beta}-catenin/CBP signaling ameliorates renal interstitial fibrosis. J Am Soc Nephrol 22:2011;1642–1653.
crossref pmid pmc
100. Henderson WR Jr, Chi EY, Ye X, Nguyen C, Tien YT, Zhou B, Borok Z, Knight DA, Kahn M. Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc Natl Acad Sci U S A 107:2010;14309–14314.
crossref pmid pmc
101. Mann J, Mann DA. Epigenetic regulation of wound healing and fibrosis. Curr Opin Rheumatol 25:2013;101–107.
crossref pmid
102. Putta S, Lanting L, Sun G, Lawson G, Kato M, Natarajan R. Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J Am Soc Nephrol 23:2012;458–469.
crossref pmid pmc
Figure. 1
Structure of the glomerular filtration barrier. (A) Electron micrograph of a normal rat glomerulus showing the basic structures of the filtration barrier. (B) A rat glomerulus 7 days after exposure in vivo to an adenovirus expressing active transforming growth factor-beta (TGF-β) demonstrates podocyte de-differentiation and foot process effacement which results in proteinuria. An adenovirus expressing active TGF-β was infused via the left renal artery in Sprague Dawley rats. Gene expression was confirmed by measuring urinary TGF-β protein. Foot process effacement was quantified, as was expression of nephrin and synaptopodin. These features suggested podocyte dedifferentiation [37].
KRCP-032-01-3-g1.jpg
Figure. 2
Effect of transforming growth factor-beta (TGF-β) on the glomerular filtration barrier. The glomerular filtration barrier consists of podocytes with their intracellular slit diaphragm proteins, the glomerular basement membrane (GBM), and the endothelial cells with associated glycocalyx. Podocytes bind to the GBM through integrins. Podocytes express vascular growth factors such as vascular endothelial growth factor (VEGF) and angiopoietins (ANGPT). Podocyte-derived VEGF and ANGPT1 act on endothelial cells to maintain cellular health and an intact filtration barrier. TGF-β is produced by endothelial cells in response to increased VEGF, or is derived from mesangial cells exposed to injury such as high concentration of glucose. TGF-β affects all structures of the filtration barrier including the GBM where it induces thickening and GBM matrix production. TGF-β induces an EMT-like phenomenon in the podocyte with loss of slit diaphragm proteins such as nephrin, and increased expression of mesenchymal proteins. We have demonstrated a role for TGF-β in the induction of ANGPT1 and ANGPT2 in podocytes, with an increase in the ANGPT receptor tyrosine endothelial kinase TEK that is responsible for some of the TGF-β-induced podocyte changes.
KRCP-032-01-3-g2.jpg
TOOLS
METRICS Graph View
  • 25 Crossref
  • 27 Scopus
  • 1,449 View
  • 14 Download
Related articles


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
#301, (Miseung Bldg.) 23, Apgujenog-ro 30-gil, Gangnam-gu, Seoul 06022, Korea
Tel: +82-2-3486-8736    Fax: +82-2-3486-8737    E-mail: registry@ksn.or.kr                

Copyright © 2024 by The Korean Society of Nephrology.

Developed in M2PI

Close layer