33
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Propyl Gallate Plays a Nephroprotective Role in Early Stage of Diabetic Nephropathy Associated with Suppression of Glomerular Endothelial Cell Proliferation and Angiogenesis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          There is growing evidence suggesting that glomerular endothelial cell proliferation and angiogenesis may be responsible for the pathophysiological events in the early stage of diabetic nephropathy. This study was designed to investigate the factors related to glomerular endothelial cell proliferation and glomerular angiogenesis and assess the effect of propyl gallate on preventing these disorders in diabetic rats. We found that glomerular hypertrophy, glomerular mesangial matrix expansion, and albuminuria were significantly increased in DN rats. CD31+ endothelial cells significantly increased in glomerulus of diabetic rats. Double immunofluorescence staining showed some structurally defective vasculus tubes in glomerulus. Real-time PCR and western blot demonstrated the glomerular eNOS expression remained at the same level, while remarkable decreased NO productions and suppressed eNOS activities were observed in diabetic rats. Treatment with propyl gallate improved glomerular pathological changes, reduced endothelial cell proliferation, decreased albuminuria, and restored eNOS activity, but did not alter eNOS expression. These data suggest that endothelial cell proliferation and immature angiogenesis may be the contributors to progression of DN. Propyl gallate is a potential novel therapeutic agent on prevention of diabetic nephropathy.

          Related collections

          Most cited references33

          • Record: found
          • Abstract: found
          • Article: not found

          Hyperglycemia inhibits endothelial nitric oxide synthase activity by posttranslational modification at the Akt site.

          Endothelial nitric oxide synthase (eNOS) is activated by phosphorylation of serine 1177 by the protein kinase Akt/PKB. Since hyperglycemia-induced mitochondrial superoxide overproduction increases O-linked N-acetylglucosamine modification and decreases O-linked phosphorylation of the transcription factor Sp1, the effect of hyperglycemia and the hexosamine pathway on eNOS was evaluated. In bovine aortic endothelial cells, hyperglycemia inhibited eNOS activity 67%, and treatment with glucosamine had a similar effect. Hyperglycemia-associated inhibition of eNOS was accompanied by a twofold increase in O-linked N-acetylglucosamine modification of eNOS and a reciprocal decrease in O-linked serine phosphorylation at residue 1177. Both the inhibition of eNOS and the changes in its post-translational modifications were reversed by antisense inhibition of glutamine:fructose-6-phosphate amidotransferase, the rate-limiting enzyme of the hexosamine pathway, or by blocking mitochondrial superoxide overproduction with uncoupling protein-1 (UCP-1) or manganese superoxide dismutase (MnSOD). Immunoblot analysis of cells expressing myc-tagged wild-type human eNOS confirmed the reciprocal increase in O-linked N-acetylglucosamine and decrease in O-linked serine 1177 phosphorylation in response to hyperglycemia. In contrast, when myc-tagged human eNOS carried a mutation at the Akt phosphorylation site (Ser1177), O-linked N-acetylglucosamine modification was unchanged by hyperglycemia and phospho-eNOS was undetectable. Similar changes in eNOS activity and covalent modification were found in aortae from diabetic animals. Chronic impairment of eNOS activity by this mechanism may partly explain the accelerated atherosclerosis of diabetes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Abnormal Angiogenesis in Diabetic Nephropathy

            Diabetic retinopathy is the leading cause of blindness in the Western world (1) and is characterized by abnormal angiogenesis driven by several factors, including tissue ischemia and hyperglycemia. This abnormal angiogenesis results in new vessels that are often immature and play a pathological role in retinopathy, contributing to both vitreous hemorrhage and fibrosis (2). In addition, increased vascular permeability leading to plasma leakage accounts for the development of macula edema, disrupting visual function (2). These evidences have led to the development of several therapeutic strategies targeting angiogenesis in diabetic retinopathy (3). Abnormal angiogenesis also occurs in diabetic nephropathy; therefore, the overriding question is whether new vessel formation in the kidney plays a pathological role in diabetic nephropathy similar to that observed in retinopathy. Intriguingly, the progression of both diabetic retinopathy and nephropathy is altered by vascular growth factor signaling through receptor tyrosine kinases, specifically involving the vascular endothelial growth factor (VEGF)-A and angiopoietin families. This review discusses abnormal angiogenesis and the role of both VEGF-A and angiopoietins in diabetic nephropathy. Evidence of abnormal angiogenesis in diabetic nephropathy. In 1987, Osterby and Nyberg (4) described abnormal blood vessels in glomeruli of patients with long-term type 1 diabetes, and later these findings were shown to occur in type 2 diabetic patients (5,6) (Fig. 1 A). The abnormal vessels occupied 1–5% of glomerular capillary area, they were occasionally dilated, and the glomerular basement membrane adjacent to them was found to be focally extremely thin. Abnormal vessels were also present in Bowman's capsule or in the glomerular vascular pole, the latter of which could often be detected as an “extra efferent arteriole” (4,7). Min and Yamanaka (8) then performed detailed analyses of computer-generated three-dimensional images in 94 patients with diabetic nephropathy and found the presence of extravessels. Intriguingly, in this study the abnormal vessels anastomosed to the lobular structure of the intraglomerular capillary network, mainly to afferent branches through the widened vascular hilus, while the distal end of the vessels connected to the peritubular capillary. In these vessels, native endothelial cell function was likely impaired, with the endothelial cells initially swollen and endothelial thickness gradually decreasing as diabetes progressed (9,10). It was also documented that the vascular wall was thickened, owing to an accumulation of matrix in these arterioles (10). Of importance was the finding that these vessels were observed in diabetic patients during the first 2 years of disease (8), which supports the contention that the development of these vessels occurs even in the early phases of diabetic nephropathy. FIG. 1. Abnormal angiogenesis in diabetic nephropathy. A: Extraglomerular neovascularization (black arrows) are found in type 2 diabetic patients. Reprinted with permission from ref. 6. B: Similarly, immunohistochemistry for CD34, a marker for endothelial cells, indicates the normal glomerular capillary pattern (brown) in nondiabetic C57BL6 mice (a). Alternatively, abnormal capillary formation is observed around glomerulus in diabetic mice lacking endothelial nitric oxide synthase (b), resembling abnormal angiogenesis in human diabetic nephropathy. Reprinted with permission from ref. 33. C: The association of abnormal angiogenesis with VEGF expression in diabetic eNOSKO mice. In contrast to glomerulus in normal mice, glomerular endothelial staining is increased along with VEGF expression. However, hydralazine treatment attenuates the increase in glomerular capillary number and VEGF expression in diabetic eNOSKO mice. D: Glomerular filtrate and atubular glomerulus in diabetic eNOSKO mice. Glomerular filtrates are delivered to the outside of the glomerulus (arrow in a) and spread to the glomerulotubular junction (arrows in a and b). This filtrate may lead to a disconnection between glomerulus and proximal tubules (b). Ballooning of Bowman's capsule is observed in c. Glomeurlar filtrate can be observed outside of Bowman's capsule in the tubular pole, where the proximal tubulus is completely disconnected from the glomerulus. Bar = 40 μm. (A high-quality digital representation of this figure is available in the online issue.) In diabetic animals, Nyengaard and Rasch (11) identified abnormal glomerular capillaries in an animal rat model induced by streptozotocin. They determined that after both 10 and 50 days following injection, the average total surface area, length, and numbers of glomerular capillaries were elevated compared with those of controls. Similarly, db/db mice were found to exhibit increased endothelial cell number and elongation of capillaries in their glomeruli (12,13). Recently, the occurrence of excessive blood vessel formation in diabetes has been demonstrated by immunohistochemistry using endothelial cell markers. As shown in Fig. 1 B, endothelial cell staining was increased in streptozotocin-induced diabetic animals (13 –16). However, it should be noted that the later stages of diabetic nephropathy are accompanied by capillary loss and rarefaction in both humans and animal models, a concept that is discussed below (5,15,17). The pathological role of abnormal angiogenesis in diabetic nephropathy. While the pathological role of abnormal vessels remains unclear, it has been demonstrated that neovascularization is associated with glomerular hypertrophy in diabetic nephropathy. Morphological changes in capillaries such as elongation and increased number contribute to glomerular hypertrophy in both humans and animals with diabetes, whereas changes in mean capillary diameter do not correlate with alterations in glomerular volume (7,12). Interestingly, the development of abnormal vessels was observed in the extraglomerular area and associated with glomerular hypertrophy in both diabetic animals and patients (6,10). Osterby et al. (7) performed a series of studies using electron microscopy and found that abnormal vessels in the vascular pole were associated with enhanced glomerular hypertrophy and increased frequency of glomerular capillary occlusion, fibrinoid lesions, tubulointerstitial injury, and urinary albumin excretion (6,10,18). Additionally, recent evidence has indicated that blocking angiogenesis attenuated glomerular basement membrane thickening, mesangial expansion, and transforming growth factor (TGF)-β1 expression in diabetic animals (13,14,16), suggesting that these vessels have a causal role in the development of early features of diabetic nephropathy. The abnormal additional vessels found in diabetes possess a thin wall at the basement membrane, while endothelial cells are swollen, suggesting that they are structurally immature and capable of causing an increase in vascular permeability (4,9,19). An increase in capillary permeability often results in the extravasation of plasma protein as well as the forming of lesions in diabetic nephropathy. For example, the capsular drop, fibrin cap, and arteriolar hyalinosis, all of which characterize human diabetic nephropathy, can be considered a consequence of plasma leakage (20). Furthermore, recent studies documented that leakage of plasma protein results in the development of atubular glomeruli in type 1 diabetic patients (21). A potential mechanism for this observation could be due to the glomerular and peritubular filtrate spreading into the glomerulo-tubular junction to eventually separate proximal tubules from the glomerulus (22). Given these facts, the extravasations from immature vessels could have a causal role in the development of diabetic nephropathy. Mechanisms for the development of abnormal angiogenesis in the diabetic kidney. Angiogenesis is often associated with an increase in endothelial number caused by an imbalance in cell proliferation and apoptosis. Recently, Hohenstein et al. demonstrated that in type 2 diabetic patients, an increased endothelial number was observed and early glomerular lesions were caused by a combination of increased proliferation and decreased apoptosis in glomerular endothelial cells (5). A major driver in this process appears to be VEGF-A expression, which is induced by high glucose levels in the early phases of diabetes and can stimulate endothelial cell proliferation and inhibit apoptosis. In addition, high glucose levels alone can enhance endothelial cell proliferation (23). Therefore, the beneficial effect of insulin treatment to block the progression of extravessels in patients with type 1 diabetes (7) could be attributed to reduced blood glucose levels and inhibition of VEGF-A expression (15). Glomerular hypertension may be another important driver in the progression of abnormal angiogenesis in diabetes. Osterby et al. (7) demonstrated that treatment with ACE inhibitors or β-blockers for 8 years to reduce hypertension in diabetic patients suppressed progression of glomerular lesions and extravessel formation. In a similar fashion, we found that lowering blood pressure in a novel animal model of diabetic nephropathy using endothelial nitric oxide synthase (eNOS) knockout (eNOSKO) mice and steptozotocin injection led to attenuated progression of abnormal angiogenesis. These mice developed abnormal vessels accompanied by advanced lesions including nodular lesions and mesangiolysis (15). As shown in Fig. 1 C, lowering blood pressure in these animals using hydralazine blocked the development of abnormal angiogenesis and inhibited glomerular VEGF-A expression (24). These data suggest that the beneficial effect of lowering blood pressure could be mediated by VEGF-A inhibition. Alternatively, one could postulate that these vessels function as a by-pass to reduce intraglomerular pressure given that abnormal vessels were found to connect intraglomerular capillaries to peritubular capillaries (8). Hence, reducing intraglomerular pressure as a consequence of lowering systemic blood pressure might reduce the need for the development of by-pass vessels. A depiction of the factors affecting abnormal angiogenesis and their pathological effects is shown in Fig. 2. FIG. 2. Mechanism and pathogenic role of abnormal angiogenesis in diabetic nephropathy. VEGF as a mediator of abnormal angiogenesis in diabetic nephropathy. The VEGF-A family has a role in the development, maintenance, and remodeling of the vasculature, acting through the receptor tyrosine kinases VEGFR-1 and VEGFR-2 (25). The VEGF-A family is very complex with several isoforms generated by alternative splicing of exons 6 and 7. In diabetes, the VEGF-A164 and VEGF-A188 isoforms are increased and can be reduced by insulin treatment (26). Additional isoforms with anti-angiogenic properties termed VEGF-Axxxb occur due to exon 8 distal splice site selection (25), leading to an unique carboxy-terminal sequence. Several studies have examined the expression pattern of the VEGF-A family in diabetic animals and patients. Cooper et al. (27) examined VEGF-A and VEGFR-2 in short- and long-term diabetic rats (3 and 32 weeks following streptozotocin injection, respectively). Short-term diabetes led to elevated VEGF-A and VEGFR-2 mRNA, whereas in long-term diabetic animals VEGF-A remained elevated and VEGFR-2 was unaltered. VEGF-A was localized to podocytes and, to a lesser extent, tubular epithelial cells, whereas VEGFR-2 was expressed in glomerular and peritubular capillaries. Elevated VEGF-A has been confirmed in our animal model of diabetic nephropathy using eNOSKO mice (15). The mice developed excessive vessels in glomeruli and tubulointerstitium that were associated with upregulation of glomerular VEGF-A expression. Elevation of VEGF-A has also been observed in human biopsy samples where the number of extravessels around the glomerular vascular pole was associated with upregulation of VEGF-A expression in the kidney (6). Finally, examination of urinary VEGF-A showed significant elevations in type 2 diabetic patients (28), compared with healthy control subjects, that positively correlated with urinary albumin-to-creatinine ratio and negatively correlated with creatinine clearance. A potential consequence of high levels of VEGF-A will be enhanced vascular permeability in the glomerulus (29). In addition, low NO (nitric oxide) bioavailability observed in diabetes (30,31) could be an additional contributor to the increased vascular permeability. Predescu et al. (32) documented that low levels of endothelial-derived NO altered the integrity of interendothelial junctions in capillaries, resulting in an increase in vascular permeability. As such, a low NO bioavailability along with high VEGF-A expression (we term this condition “uncoupling of VEGF-A with NO”) observed in the diabetic milieu of eNOSKO mice could potentiate the vascular permeability in the glomerulus and cause glomerular injury in diabetic nephropathy (15,33). Intriguingly, this uncoupling condition could also cause the development of abnormal angiogenesis. This notion can be supported by recent evidences from our laboratory and other groups that NO can negatively regulate VEGF-A–induced endothelial proliferation (34), whereas NO deficiency enhances VEGF-A activity, leading to endothelial proliferation (35). We have extensively reviewed a causal role of this uncoupling condition in other types of vascular diseases, including coronary artery disease, remnant kidney, and angiotensin II–induced renal injury in previous work (33). It should be noted that while the aforementioned studies indicate low NO contributing to capillary hyperpermeability, Tilton et al. (36) demonstrated that supra-physiological NO positively mediates hyperpermeability in response to exogenous VEGF-A in several different nondiabetic tissues. Therefore, it is likely that physiological levels of NO are required to maintain low vascular permeability and that NO levels that are either too high or too low (depending on the biological situation) may lead to hyperpermeability. VEGF-A is lowered in the advanced stage of diabetic nephropathy. The study by Cooper et al. (27) suggested that although VEGF-A may be elevated in the initial phases of diabetic nephropathy, it may not be maintained as more chronic fibrotic changes occur in the kidney. Indeed, in many animal models of chronic kidney disease, VEGF-A levels are reduced, correlating with the progression of renal damage (37,38). To examine this in diabetic nephropathy, Baelde et al. (17) used laser-capture microdissection to determine gene expression in glomeruli from 28 diabetic patients. They observed a reduction of 2.5-fold in VEGF-A expression in severely injured glomeruli as evidenced by a loss of endothelial cells and a reduction in podocyte markers (WT-1, nephrin, and podocin mRNAs) (17). Given that podocytes and tubular epithelial cells are the primary source of VEGF-A in the kidney, the mechanism for a reduction in VEGF-A expression in severe renal injury could be attributed to the inability of these cells to produce VEGF-A due to advanced stages of cellular injury. Other studies have found that VEGF-A expression was decreased in sclerotic areas and in nodular lesions of diabetic nephropathy (39,40). In addition, Zucker diabetic fatty rats exhibited a decline in renal VEGF-A expression in advanced stages of diabetic nephropathy (41,42). This interesting concept was highlighted in an elegant study by Hohenstein et al. (5) where they used specific antibodies to examine not only VEGF-A expression but also receptor-bound VEGF-A as a marker of bioactivity in diabetic patients. In their study, although VEGF-A expression was increased in all diabetic glomeruli by many cell types, VEGF-A activity was only increased in the endothelium of mildly injured glomeruli and significantly decreased in more severe glomeruli (5). This data suggests that the upregulation of VEGF-A in early stages of diabetic nephropathy may provide a mechanism for the initial progression of the disease, leading to excessive blood vessel formation. The decline of VEGF-A in the later phase of diabetic nephropathy may reflect a loss of endogenous VEGF-A due to the disruption of podocytes and tubular cells in chronic kidney damage (Table 1). TABLE 1 Reduction of VEGF expression in diabetic nephropathy Renal VEGF expression Endothelium Diabetes history Stage of nephropathy Ref. Human Decreased (mRNA, protein) NE Not mentioned Sclerotic glomerulus 40 Human Decreased (mRNA) NE Sclerotic glomerulus with heavy proteinuria 70 Human type 2 diabetes Decreased (mRNA) NE >5 years Glomerulosclerosis (24–33%) and tubulointerstitial injury (25–50%) 71 Human type 2 diabetes Decreased mRNA in VEGF165, increased mRNA in VEGF121 NE >2 years Microalbuminuria, 10 of 17 patients; macroalbuminuria, 7 of 17 patients 72 Human type 2 diabetes Decreased (protein) Decreased EC proliferation, increased EC apoptosis 12.8 ± 7.8 years Advanced lesions 5 Human type 2 diabetes Decreased (mRNA, protein) Decreased CD31 (+) EC >5 years Interstitial fibrosis, podocyte loss 17 Human type 1 and 2 diabetes Decreased (mRNA, protein) Decreased CD31 (+) EC Established diabetes or early diabetes EC loss in peritubular capillary or tubulointerstitial injury 73 Zucker rat Decreased (mRNA, protein) NE 22 weeks old Advanced renal injury 41 EC, endothelial cell; NE, not examined. Alterations in angiopoietin balance as a molecular mechanism of diabetic nephropathy. A second family of growth factors implicated in the progression of diabetic nephropathy are the angiopoietins, which are critical for the normal vascular differentiation, maintenance, and turnover of blood vessels in mature animals (43). Angiopoietin-1 and -2 are ligands for the Tie-2 receptor tyrosine kinase, expressed mainly by endothelia; angiopoietin-1 stimulates receptor activation, leading to promotion of endothelial survival and stabilization. Angiopoietin-2 is considered a natural antagonist of angiopoietin-1 (44), although other data suggest that high concentrations of angiopoietin-2 may activate Tie-2 (45). Alterations in the expression of the angiopoietins have been implicated in the progression of diabetic nephropathy (rev. in (43). In addition, transgenic mice with inducible overexpression of angiopoietin-2 in podocytes in otherwise normal healthy adult animals develop significant increases in albuminuria (46), a parameter that correlates with, and can predict, the progression of renal damage in diabetes (47). Collectively, these observations suggest that a decreased ratio of angiopoietin-1 to angiopoietin-2 might play a role alongside VEGF-A in the pathobiology of diabetic nephrology. Importantly, the biological effects of angiopoietin-2 are context dependent and, in vivo, depend on ambient levels of VEGF-A, such that vessel regression occurs if VEGF-A is lacking, whereas vessel destabilization followed by angiogenesis occurs if the local milieu is rich in VEGF-A (44). It could be postulated that the increased levels of angiopoietin-2 alongside a VEGF-A–rich milieu in glomeruli during the initial phases of diabetes will lead to the destabilization of blood vessels and hence excessive angiogenesis. Therefore, it is possible that modulation of the balance between angiopoietin-1 and -2 may have therapeutic potential in diabetic nephropathy. Targeting angiogenesis to treat diabetic nephropathy. Given the evidence above, there is a rationale for targeting angiogenic pathways to prevent diabetic nephropathy, and several studies have now blocked VEGF-A activity as a therapy to prevent abnormal angiogenesis. An elegant genetic approach was recently taken by Gnudi and colleagues (48) by blocking VEGF-A signaling in mice administered streptozotocin through overexpressing soluble VEGFR-1, specifically in podocytes. Diabetic mice that overexpressed soluble VEGFR-1 had attenuated albumin excretion, mesangial expansion, glomerular basement membrane thickening, podocyte foot process fusion, and TGF-β1 expression (48). de Vriese's group (49) examined the effect of treatment with a monoclonal anti–VEGF-A antibody in the early phase of diabetes induced by streptozotocin. Administration of the antibody decreased hyperfiltration, albuminuria, and glomerular hypertrophy in diabetic rats. Although the effect on angiogenesis was not specifically examined in this study, VEGF-A blockade prevented the upregulation of eNOS associated with this model (49). Other studies were performed in db/db mice and the Zucker diabetic fatty rat (50,51). In db/db mice, VEGF-A antibody treatment resulted in a reduction in kidney weight, glomerular volume, basement membrane thickness, and urinary albumin excretion (50); in the Zucker diabetic fatty rat, VEGF-A antibody treatment prevented glomerular hypertrophy. However, neither of these studies examined the effect of reducing VEGF-A on abnormal angiogenesis. Similarly, Sung et al. (52) blocked the phosphorylation of the VEGF-A receptors using the pharmacological kinase inhibitor SU5416 in db/db mice and found that this approach prevented the development of albuminuria and glomerular basement membrane thickening. Interestingly, blocking VEGF-A activation prevented the loss of nephrin and improved structural changes in podocyte foot processes in db/db mice. These results suggest that VEGF-A could impair podocyte function, which may be an additional mechanism by which VEGF-A causes urinary protein excretion. However, since these studies did not examine the process of abnormal angiogenesis per se, further experiments are required to determine whether this beneficial effect of anti–VEGF-A therapy could be due to the blocking of VEGF-associated angiogenesis. Currently VEGF-A inhibitors are classified into four groups (Table 2) and have been used in clinical practice. Importantly, the efficacy of these individual compounds is not identical. For instance, the tyrosine kinase inhibitors have greater anti-tumor efficacy only at early stages of cancer progression (53), whereas monoclonal antibodies are capable of regressing tumor growth (54). In the kidney, VEGF-A function is also complicated given that it has been found to exhibit both deleterious and beneficial effects (rev. in (33). In fact, VEGF-A is found to be deleterious in diabetic nephropathy but largely beneficial in nondiabetic animal models of renal disease. Hence, we need to be cautious before using VEGF-A inhibitors in the diseased kidney. Previously, the beneficial effect of anti–VEGF-antibodies was shown in two diabetic animal models: streptozotocin-induced diabetic rats and db/db mice (49,55). On the contrary, it has been postulated that a potential adverse effect with VEGF-A inhibitors could be endothelial injury because endothelial cells require VEGF-A in physiological conditions. Eremina et al. (56) demonstrated that bevacizumab, the anti–VEGF-antibody, causes renal thrombotic microangiopathy partly due to endothelial injury in patients. Similarly, Advani et al. (57) demonstrated that VEGFR-2 tyrosine kinase inhibitors exacerbated hypertension and renal disease in hypertensive rats. Likewise, systemic overexpression of soluble VEGFR-1 in normal animals was found to cause endotheliosis and podocyte injury, leading to proteinuria and hypertension (58,59). In addition, the deleterious effect of anti-VEGF antibodies could be attributed to the deposition of VEGF–anti-VEGF complex, C3 deposition, and endothelial swelling (54). However, in some experiments, it was also shown that normal kidneys did not have any side effects from VEGF inhibitors treatment (60,61). TABLE 2 VEGF inhibitors Function Drug name Inhibition of VEGF secretion Iressa* Tarceva* Sequestration of VEGF Bevacizumab* Ranibizumab* Pegaptanib* Blocking binding of VEGF to VEGFR DC101 Inhibitior of receptor tyrosine kinase Sunitinib* Sorafenib* *Approved by the U.S. Food and Drug Administration. Since endothelial cells require VEGF-A in physiological conditions, substantial inhibition can cause endothelial injury. In this regard, it may not be adequate to use VEGF-A inhibitors in patients with normal kidney function or in nondiabetic renal injury in which VEGF-A expression is downregulated. In contrast, Gnudi and colleagues (48) succeeded in treating diabetic nephropathy using podocyte-specific overexpression of soluble VEGFR-1. In this study, neither VEGF-A expression nor VEGFR-2 phosphorylation was significantly blocked by overexpression of soluble VEGFR-1 in the diabetic kidney, suggesting that VEGF-A function was partially inhibited. Thus, the “partial” inhibition might be a means to treat diabetic nephropathy without any adverse effects. Further clarification on the adverse effects of VEGF-A inhibitors is required before they may be used to treat diabetic patients. Angiopoietins have been used therapeutically in several diabetes situations. Administration of angiopoietin-1 has been shown to suppress diabetic retinopathy by preventing leukocyte adhesion, endothelial cell injury, and blood-retinal barrier breakdown (62). With regard to diabetic nephropathy, Lee et al. (63) demonstrated that systemic adenoviral delivery of COMP-Ang-1 (a modified form of angiopoietin-1) reduced renal fibrosis in db/db mice. However, this strategy also caused a significant improvement in hyperglycemia, an event possibly related to the systemic administration of angiopoietin-1, which could itself, at least partly, account for the amelioration of diabetic nephropathy. Therefore, further experiments are required to examine whether modulation of this pathway could be a future treatment for patients with diabetic nephropathy. Several studies have attempted to block angiogenesis using other anti-angiogenic molecules in animal models, as shown in Table 3 (13,14,16,64). Angiostatin is a potent angiogenic inhibitor that blocks proliferation, induces apoptosis, and prevents migration of endothelial cells in vitro. In addition, angiostatin has anti-inflammatory actions by inhibiting leukocyte recruitment and both neutrophil and macrophage migration. In streptozotocin-induced diabetic nephropathy, adenoviral-mediated delivery of angiostatin was found to alleviate albuminuria and glomerular hypertrophy (64). We also found a similar advantage of angiostatin treatment in the remnant kidney model (65). Similarly, endostatin, a potent inhibitor of angiogenesis derived from type XVIII collagen (14), and tumstatin, an angiogenic inhibitor derived from type IV collagen (16), were both able to prevent glomerular hypertrophy, hyperfiltration, and albuminuria in type 1 diabetic mice. Interestingly, these treatments were shown to prevent mesangial expansion and inflammation and also to attenuate the increase in levels of VEGF-A and angiopoietin-2 normally observed in this model (14,16) independent of blood pressure and blood glucose levels (Fig. 2). Similar observations were made with 2-(8-hydroxy-6-methoxy-1-oxo1H-2-benzopyran-3-yl) propionic acid (a small molecule with anti-angiogenic activity) in db/db mice (13). These novel treatments to prevent angiogenesis could be considered for patients in early stages of diabetic nephropathy. TABLE 3 Angit-angiogenic therapy in diabetic nephropathy Anti-VEGF antibody SU5416 Angiostatin Endostatin Tumstatin NM-3 PEDF Blocking VEGF Pan-VEGFR kinase inhibitor Proteolytic fragment of plasminogen NC1 domain of type 18 collagen NC domain of the α3 chain of type 4 collagen 2-(8-hydroxy-6-methoxy-1-oxo-1H-2-benxopyran-3-yl) propionic acid Pigment epithelium-derived factor STZ Wister rat* db/db mouse* GK rat* db/db mouse STZ Brown Norway rat STZ-C57BL6 mouse* STZ-C57BL6 mouse* db/db mouse* STZ Brown Norway rat Age or weight 250–280 g 8 weeks 8 weeks 8 weeks 8 weeks 7–8 weeks 7–8 weeks 7–8 weeks 8 weeks Treatment duration ≤6 weeks 60 days 6 weeks 8 weeks 2–3 weeks 4 weeks 2–3 weeks 8 weeks 3 weeks Increase in CD31(+) endothelial cell in glomeruli NE NE NE NE NE Blocked Blocked Blocked NE Renal hypertrophy NE Blocked NE NE NE Blocked Blocked Not blocked NE Glomerular hypertrophy Blocked Blocked No effect NE Blocked Blocked Blocked Blocked NE Mesangial expansion NE Tended to be lowered NE Blocked NE Blocked Blocked Blocked NE Glomerular basement membrane thickening NE Blocked NE Blocked NE NE NE NE NE Hyperfiltration Blocked Blocked No effect NE NE Blocked Blocked Blocked NE Urinary albumin Decreased Decreased No effect Decreased Decreased Decreased Decreased Decreased Decreased Podocyte injury or Nephrin expression NE NE NE Improved NE Recovered (Nephrin expression) Recovered (Nephrin expression) Recovered (Nephrin expression) NE Macrophage infiltration NE NE NE NE NE Blocked Blocked Blocked NE VEGF expression NE NE NE NE Decreased Decreased Decreased Decreased NE TGF-β1 expression NE NE NE NE Decreased Decreased NE Decreased Decreased Reference 49 50 51 52 64 14 16 13 74, 75 NE, not examined; STZ, streptocotocin. *Female. What prospects are there for other novel therapies for diabetic nephropathy? One area of interest may be in examining the anti-angiogenic isoforms of VEGF-Axxxb in models of diabetic nephropathy, which may open new avenues of treatment strategies. Another therapy could be the use of RNA aptamers, which are oligonucleotidue ligands that bind with high-affinity to molecular targets. One such aptamer that targets VEGF-A165 has been used successfully in clinical trials to block ocular neovascularization (66). Promising results have also been obtained using small-molecule tyrosine kinase inhibitors to treat type 1 diabetic mice (67); however, the kidneys were not examined in these studies. Finally, other molecules involved in angiogenic pathways such as the Notch family (68) may provide interesting information in the pathobiology and treatment of diabetic nephropathy. In this regard, studies by Niranjan et al. (69) have already demonstrated that lack of the Notch1 transcriptional partner Rbpj in podocytes is able to modulate the progression of albuminuria in diabetic mice. In conclusion, while the presence of abnormal angiogenesis was demonstrated more than a decade ago, we are only beginning to unravel the pathophysiological importance of this event. Anti-angiogenic treatments can prevent the progression of animal models of diabetic nephropathy, but further studies are required before these treatments can be used in a clinical setting. The fact that diabetic nephropathy is currently still the leading cause of end-stage renal disease points to the need for additional treatment strategies. Thus, novel therapies that target other angiogenic pathways such as the angiopoietin and Notch families could be an attractive option to block diabetic nephropathy in the future.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              NAD(P)H oxidase and uncoupled nitric oxide synthase are major sources of glomerular superoxide in rats with experimental diabetic nephropathy.

              Increased production of reactive oxygen species (ROS) in diabetes may be a common pathway linking diverse pathogenic mechanisms of diabetic vascular complications, including nephropathy. Assessment of the oxidative stress production pathway is therefore important for the prediction and prevention of diabetic complications. However, ROS production mechanisms remain unclear in diabetic glomeruli. To identify the source and determine the mechanisms of ROS production in the diabetic kidney, diabetes was induced with streptozotocin in rats. After 6 wk, glomerular ROS production had increased in the streptozotocin rat kidney, as assessed by dihydroethidium-derived chemiluminescence. ROS production was increased by the addition of NADH or L-arginine and was partially reduced by the addition of diphenylene iodonium or N(G)-nitro-L-arginine methyl ester, identifying NAD(P)H oxidase and nitric oxide (NO) synthase (NOS) as ROS sources. The mRNA and protein expression of endothelial NOS (eNOS), as measured by real-time RT-PCR and Western blotting, increased significantly (mRNA level, 1.3-fold; protein level, 1.8-fold). However, the dimeric form of eNOS was decreased in diabetic glomeruli, as measured by low-temperature SDS-PAGE. Production of renal ROS and NO by uncoupled NOS was imaged by confocal laser microscopy after renal perfusion of 2',7'-dichlorofluorescein diacetate (a ROS marker) and diaminorhodamine-4M AM (a NO marker) with L-arginine. Accelerated ROS production and diminished bioavailable NO caused by NOS uncoupling were noted in the diabetic kidney. Administration of tetrahydrobiopterin (BH4), a cofactor for eNOS, reversed the decreased dimeric form of eNOS and glomerular NO production. Our results indicate that NAD(P)H oxidase and uncoupling of eNOS contribute to glomerular ROS production, mediated by the loss of BH4 availability. These mechanisms are potential key targets for therapeutic interventions.
                Bookmark

                Author and article information

                Journal
                Exp Diabetes Res
                Exp Diabetes Res
                EDR
                Experimental Diabetes Research
                Hindawi Publishing Corporation
                1687-5214
                1687-5303
                2012
                4 September 2012
                : 2012
                : 209567
                Affiliations
                1Department of Nephrology, Renmin Hospital, Hubei University of Medicine, Hubei, Shiyan 442000, China
                2Clinic Medicine Research Institute, Renmin Hospital, Hubei University of Medicine, Hubei, Shiyan 442000, China
                3Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, Shiyan 442000, China
                Author notes

                Academic Editor: A. Veves

                Article
                10.1155/2012/209567
                3439983
                22988451
                03df9ab6-d4cf-4159-b891-3ee33597ab39
                Copyright © 2012 Shaojiang Tian et al.

                This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 18 May 2012
                : 9 July 2012
                : 10 July 2012
                Categories
                Research Article

                Endocrinology & Diabetes
                Endocrinology & Diabetes

                Comments

                Comment on this article