Role of VEGF in Kidney Development, Microvascular Maintenance and Pathophysiology of Renal Disease

Article information

Korean J Intern Med. 2003;18(2):65-75
Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea;
*Department of Medicine, New York Medical College, Valhalla, New York, USA
Correspondence to : Byung-Soo Kim, M.D., Our Lady of Mercy Hospital, Pupyung-dong 665, Pupyung-Gu, Incheon 403-720, Korea, Tel: 82-32-510-5683, Fax:82-32-510-5683, E-mail: kbsnep@yahoo.co.kr
Received 2003 January 28; Accepted 2003 February 28.

Abstract

Vascular endothelial growth factor, VEGF, is essential for endothelial cell differentiation (vasculogenesis) and for the sprouting of new capillaries from preexisting vessels (angiogenesis). In addition, there is strong evidence that VEGF is a survival factor allowing the cells to survive and proliferate under conditions of extreme stress.

Hypoxia is a key regulator of VEGF gene expression. Besides hypoxia, many cytokines, hormones and growth factors can up-regulate VEGF mRNA expression in various cell types.

VEGF is present in the glomerulus of both the fetal and adult kidney. The VEGF produced by glomerular epithelial cell may be responsible for maintenance of the fenestrated phenotype of glomerular epithelial cells, thus facilitating the high rate of glomerular ultrafiltration. But there is little known about the role of VEGF in the tubule.

VEGF is thought to be involved in many kinds of kidney diseases. Whereas VEGF has a beneficial role in the pathogenesis in some diseases, it does harmful action in others. Because VEGF is known to be associated with the pathogenesis of some diseases, such as diabetic nephropathy, renal tumor and polycystic kidney disease, the study about the role of VEGF is going to be a target for disease control. On the other hand, an attempt at enhancing the role of VEGF has to be made at diseases like several ARF models and experimental glomerulonephritis.

Keywords: VEGF; Kidney; Disease

INTRODUCTION

Vascular endothelial growth factor, VEGF, also termed vascular permeability factor, VPF, has been at the center stage of vascular research for the past 2 decades. These years witnessed the expansion of the VEGF family of proteins, elucidation of their functions and attempts at utilizing these proteins and their respective neutralizing antibodies for therapeutic purposes, both in experimental animals and in clinical trials of myocardial and limb revascularization.

Angiogenesis is essential not only for organ development and differentiation during embryogenesis but also for physiological situations, such as wound healing and reproductive functions1). Vascular supply is also thought to be implicated in the pathogenesis of a variety of disorders, including proliferative retinopathies, age-related macular degeneration, tumors, rheumatoid arthritis and psoriasis. Vascular endothelial growth factor (VEGF) is essential for endothelial cell differentiation (vasculogenesis) and for the sprouting of new capillaries from preexisting vessels (angiogenesis)2, 3). In addition, there is also strong evidence that VEGF is a survival factor allowing the cells to survive and proliferate under conditions of extreme stress, both in vitro and in vivo48). This review will focus on the latest developments in understanding the role of VEGF in renal development, function and dysfunction.

VEGF family members and their receptors

VEGF is a distant relative of platelet-derived growth factor (PDGF) and is a member of a family of related growth factors that now includes VEGF-B, -C, -D and -E and placenta growth factor (PIGF)9). In particular, VEGF, referred to also as VEGF-A, is a major regulator of normal and abnormal angiogenesis.

Alternative splicing of human VEGF mRNA from a single gene containing eight exons10) gives rise to at least five different isoforms of 121, 145, 165, 189 and 206 amino acid residues7, 8, 11) Mouse and rat VEGF isoforms are shorter by one amino acid12, 13). VEGF121 fails to bind to heparin, while VEGF165 is a basic, heparin-binding protein. VEGF189 and VEGF206 are more basic and bind to heparin with ever greater affinity than VEGF16514). VEGF121, VEGF145 and VEGF165 are secreted and readily diffusible. In contrast, VEGF189 and VEGF206 are almost completely sequestrated in the extracellular matrix (ECM)15). Human VEGF165 is typically expressed as a 46-kDa homodimer of 23-kDa monomers and is the most abundant and the most biologically active form.

Two distant receptor tyrosine kinases (RTKs) have been identified for VEGF, VEGF-R1 (Flt-1) and VEGF-R2 (KDR/Flk-1), which share −44% amino acid homology with each other16, 17). A third receptor, VEGF-R3 (Flt-4), binds VEGF-C and -D and does not bind VEGF-A18, 19). PIGF and VEGF-B bind with high affinity only to VEGF-R1 and do not bind to VEGF-R2. VEGF-E binds with high affinity to VEGF- R2 (Table 1).

Binding of VEGF family members to the known VEGF tyrosine kinase receptors

Neuropilin-1 (NP-1) was recently identified as a new receptor for VEGF. NP-1, previously known as a neuronal receptor, was shown to function also in endothelial cells as an isoform-specific receptor for VEGF165 and as a co-receptor in vitro of VEGF R220).

Regulation of VEGF gene expression

Hypoxia

Hypoxia is a key regulator of VEGF gene expression, both in vitro and in vivo. Hypoxia induces transcription of the VEGF gene21, 22). In the VEGF gene, a 28 bp hypoxia response element (HRE) is located approximately 1 kb upstream of the transcription initiation site23). Transcription activation of the VEGF gene is mediated by binding of the hypoxia-inducible factor-1 (HIF-1 α/β) to this element in hypoxic condition. The HIF-1 α subunit is the pO2-sensitive partner24).

However, transcriptional activation is not the only mechanism leading to VEGF up-regulation in response to hypoxia. Increased mRNA stability is an important post- transcriptional component25, 26). Hypoxia induces transcription of VEGF gene and stabilization of VEGF mRNA.

Hormones and cytokines

Many cytokines, hormones and growth factors can up-regulate VEGF mRNA expression in various cell types. Epidermal growth factor, TGF-β or keratinocyte growth factor result in a marked induction of VEGF gene expression27). Both interleukin 1 α and prostaglandin E2 induce expression of VEGF in cultured synovial fibroblasts, suggesting the participation of such inductive mechanisms in inflammatory angiogenesis28). Insulin-like growth factor 1 has also been shown to induce VEGF mRNA and protein in cultured colorectal carcinoma cells29). Thyroid-stimulating hormones and ACTH are able to induce VEGF gene expression in vitro30, 31).

VEGF is up-regulated by renin and angiotensin II32, 33). In turn, VEGF is also known to stimulate expression of angiotensin-converting enzyme and production of angiotensin II34, 35) Although there were many reports showing that angiotensin II is a potent stimulator of VEGF expression in several cell lines32, 33, 36), one study showed the opposite relations between angiotensin II and VEGF. According to this latter study, angiotensin II is a potent inhibitor of VEGF expression in mTAL cells37).

Distribution of VEGF and its receptors in the kidney

VEGF is present in the glomerulus of both the fetal and adult kidney. By immunohistochemistry of the human kidney, VEGF is expressed in the glomerular matrix and in the glomerular cells of both the fetal and adult human kidneys38). In the adult rat kidney, VEGF is constitutively expressed in glomerular podocytes. In addition, consistent with the fact that glomerular epithelial cells (GEC) have been identified as the site of constitutive production of VEGF39), several reports showed that VEGF is present in the glomerulus of the kidney40, 41).

In the tubule, so far, VEGF expression in the normal human kidney has been known to be confined to the distal and collecting duct epithelium38). The expression of VEGF protein in the distal tubule was also found in the fetal kidney. VEGF expression in the distal tubule was weaker than in the glomerulus41).

In the rat, normal kidney showed diffuse expression of VEGF in all tubules of the renal cortex and medulla42). Proximal tubules in the human kidney exhibit only faint, if any, labeling of VEGF mRNA and VEGF protein when studied by in situ hybridization and immunohistochemistry39, 42). But, recently, some publications reported that human proximal tubular cells produced VEGF in culture43, 44).

VEGF receptors are predominantly expressed in the glomerular capillaries45). To a lesser degree, VEGF receptors are found in other capillaries and veins by immunohistochemistry. In the fetal kidney, glomeruli demonstrated strong labeling for flt-1 and KDR mRNA by in situ hybridization with antisense riboprobes38). Besides endothelial cells, other cells, such as mesangial cells46) and tubular cells47), function as effectosr of VEGF, although the precise actions on these cells remain unclear.

Functional role of VEGF in the glomerulus and tubule of the kidney

The alternative exon splicing of a single VEGF gene gives rise to 5 isoforms of VEGF. The major known functional difference among the various VEGF isoforms is their ability to bind heparin and heparan sulfate proteoglycans distributed on a cellular surface and within the extracellular matrix and basement membrane.

The fact that the glomerular epithelial cell (GEC) expresses mRNA for the soluble secretory form VEGF-120 and for the soluble matrix-associable VEGF-164 and insoluble, heparin-binding matrix-associated VEGF-188 and VEGF-205 suggests that the glomerular basement membrane may be the site of VEGF accumulation and storage. Thus we hypothesized that VEGF produced by GEC is stored at the basement membrane and released to the endothelial cells, thus effectively completing the loop of paracrine action.

VEGF is constitutively expressed in the normal kidney where it is primarily localized to glomerular podocytes and tubular cells. Furthermore, the expression of VEGF receptors on the endothelial cells as an effector system suggests that VEGF effects may be mediated via its binding to heparan sulfate proteoglycans, which act to shuttle VEGF across the basement membrane48).

It has been suggested that VEGF produced by the glomerular epithelial cell (GEC) may be responsible for maintenance of the fenestrated phenotype of glomerular epithelial cells38, 39), thus facilitating the high rate of glomerular ultrafiltration. Along these lines, we showed that VEGF acts on the permeability of endothelial cell monolayers in vitro. Caveolae, the plausible structures involved in the increase in endothelial permeability49), became organized into elongated cell-spanning structures in endothelial cells shortly after exposure to VEGF. These structures were permeable to high molecular weight dextran. Two days after addition of VEGF, endothelial cells exhibited diaphragmed fenestrae50). Thus, it is conceivable that VEGF elicits a rapid increase in vascular permeability via mobilization of caveolae and formation of cell-spanning channels, whereas the long-term effect requires formation of fenestra (Figure 1).

Figure 1.

Long-term effect of VEGF resulted in the formation of fenestration of endothelial cells. (A) HUVEC were cultured on matrigel for 36 h in the presence of 10 ng/mL VEGF-165. Light microscopic image shows an elaborate capillary-like network formed under these conditions. (B) Transmission electron micrograph shows diaphragmed fenestrae (arrowheads). (C) and (D) changes in electrical resistance and capacitance, respectively, in HUVEC treated with 10 ng/mL VEGF-165 (control cells were deprived of VEGF), demonstrating concomitant decrease in resistance and increase in capacitance compared with control. One-way ANOVA of experimental and control data in B and C showed that these curves are significantly different at p<0.05. HUVEC: human umbilical endothelial cell. Reprinted with permission from ref. 50.

In contrast to the glomerulus, little is known about the role of VEGF in the tubule. In the kidney, the peculiar anatomy of microvasculature is characterized by the first division of arterioles into glomerular capillaries, which is followed by the “rete mirabile” of the second subdivision of the efferent arterioles into peritubular capillaries. Both the glomerular and peritubular capillary endothelium are characterized by extensive fenestration and high permeability to solutes. This analogy with the microvasculature of endocrine organs and tumors provided an impetus for investigation into the production of VEGF in the different nephron segments of the kidney51).

In the remnant kidney model, Kang et al.37) observed a strong correlation between the number of peritubular capillaries and tubular VEGF expressions. Recently, we investigated the production and role of VEGF in the proximal tubular and glomerular epithelial cells by using epithelial-endothelial cell co-cultures44). Our study showed that proximal tubular epithelial cells constitutively produce VEGF and VEGF secreted by proximal tubular epithelial cells supports angiogenesis in culture. Furthermore, VEGF production is dramatically up-regulated by hypoxia and by high glucose. However, the angiogenic response in vitro is limited to the hypoxic condition only and does not occur under hyperglycemia. These data indicate that, while VEGF is necessary for angiogenesis, it is not sufficient under conditions when endothelial cells become dysfunctional, like in hyperglycemia. Collectively, we hypothesize that the regulated production of VEGF by the proximal tubular epithelium may play a profound role in the regulation of the peritubular capillary network (Figure 2).

Figure 2.

VEGF protein expression in GEC and RPTEC (A and B). Immunoprecipitation for VEGF in GEC (A) and RPTEC (B). Both bands shown are VEGF protein expressions from cell lysate and media, respectively. VEGF was detected using immunoprecipitation with polyclonal antibody followed by blotting with the monoclonal antibody. (C and D) VEGF expression in GEC (C) and RPTEC (D). GEC: rat glomerular epithelial cell, RPTEC: human renal proximal tubular epithelial cell; Con: control; D-glu: 30 mM D-glucose for 24 hours; L-glu: 30 mM L-glucose for 24 hours; Hypo: hypoxia for 24 hours; DG+H: 30 mM D-glucose+hypoxia for 24 hours.

*: p<0.01 compared with the control group, #: p<0.05 compared with the control group. Reprinted with permission from ref. 44.

VEGF in renal diseases

1. Acute renal failure

Hypoxia is a well-established potent stimulus for VEGF production in some cells, acting by inducing transcription of VEGF gene and stabilization of VEGF mRNA21, 26, 52). We also observed that VEGF production was increased after hypoxic injury in the glomerular (GEC) and human renal proximal tubular epithelial cells (RPTEC) in culture44). Interestingly, a 24-hour hypoxia resulted in a 2.4- and 4.9-fold increase in VEGF mRNA in GEC and RPTEC, respectively, indicating that VEGF production after hypoxia is more robust in the tubular cells than in the glomerulus.

The most susceptible region to hypoxia is the outer medulla53). The tubules in this region are normally in a borderline hypoxic state due to the countercurrent circulation and high oxygen demand of the medullary thick ascending tubule and the S3 segments of the proximal tubule. Thus, hypoxia can induce tubular and interstitial cell injury, cell activation, proliferation, cytokine generation and matrix synthesis associated with an increased expression of HIF-1 α54, 55).

There is strong evidence that VEGF is a survival factor for endothelial cells. VEGF acts as a survival factor by preventing endothelial cell apoptosis via stimulation of VEGF R-2-mediated signaling cascade involving PI3K/Akt pathway6, 7). This would be an obvious teleologic justification for tubular secretion of VEGF, since survival of a tubular cell during hypoxic injury is likely to be critically dependent on the survival of the adjacent endothelium and maintenance of the peritubular blood flow.

One study suggests that the change of VEGF distribution, rather than an increase in VEGF production, is responsible for the response to hypoxic injury. The authors show that acute re-distribution of cytoplasmic VEGF to the basolateral aspect of tubular epithelial cells in response to hypoxic injury42) can protect endothelial cells.

Besides ischemic injury, VEGF accelerates renal recovery in toxin-induced ARF56). VEGF resulted in greater recovery of microvasculature and better renal function in the TMA (thrombotic microangiopathy) animal model induced by the anti-glomerular endothelial cell IgG.

One study shows that peritubular capillary loss is associated with chronic tubulointerstitial injury (CTI) and the pattern of VEGF expression has been changed57). VEGF expression was increased in the tubule, especially in morphologically intact or hypertrophic ones. This supports the postulate on the role of VEGF in chronic ischemic injury.

2. Glomerular diseases

The patterns of VEGF expression in glomerular diseases are complicated. VEGF has been implicated in the induction of proteinuria in renal disease58). In addition, the up-regulation of VEGF expression in the mesangial cells during mesangio-proliferative disease was described59).

Recently, one study showed that the addition of VEGF to cultured human mesangial cells induced their proliferation and that VEGF receptors are present in human mesangial cells46). Mesangial cell proliferation is a common response of the glomerulus to diverse injuries in both human and experimental glomerular disease, and several growth factors have been implicated in this process. There are several potential sources of glomerular VEGF. VEGF can be released by glomerular cells and by infiltrating T lymphocytes60) or monocytes61). Among resident glomerular cells, epithelial39) and mesangial cells61) produce VEGF in vitro. VEGF production is up- regulated by several growth factors, inflammatory cytokines and vasoactive agents in the kidney. Thus, a series of stimuli involved in glomerular injury can induce both VEGF and modulate the expression of its receptor.

On the other hand, many studies have shown the beneficial role of VEGF in the pathogenesis of glomerular diseases. In the experimental anti-Thy-1.1 glomerulonephritis, VEGF may participate in the healing of glomerular lesions62). Another study showed that systemic administration of VEGF induced glomerular repair and resolution of glomerulonephritis in the experimentally induced glomerulonephritis63). The authors discovered that the recovery was associated with stimulation of angiogenesis, vascular remodeling and angiogenic capillary repair, all playing an important role in recovery from severe glomerular damage. Different from one report as mentioned above46), proliferation and/or activation of mesangial cells, even in rats treated with VEGF, was significantly decreased compared to the control group. The authors suggested that the dose of VEGF determined whether the proliferation and activation of mesangial cells developed. As impaired capillary regeneration in severely damaged regions relates to the continuation of mesangial cell proliferation and activation, capillary regeneration can influence the suppression of mesangial cell proliferation and activation. In addition, recent studies suggested that VEGF does not affect the development of proteinuria in renal diseases64, 65).

There are two studies suggestive of the relation between VEGF and several glomerular diseases. One study tested the urinary VEGF levels in patients with several glomerular diseases66). It appeared that normal subjects had detectable urinary VEGF excretion and, compared with them, the excretion was unchanged in minimal change disease (MCD), but was elevated in patients with FSGS and necrotizing glomerulonephritis. In contrast, MGN (membranous glomerulonephritis) was associated with the significantly suppressed urinary excretion of VEGF, while decreasing clinical activity (reduction in proteinuria) was associated with recovery of urinary VEGF excretion toward the levels observed in the healthy subjects. The other study evaluated kidney tissue from 47 patients with a variety of renal diseases using immunohistochemistry and in situ hybridization67). The authors observed that VEGF expression was decreased or absent in sclerotic glomeruli, glomeruli compressed by crescents, glomeruli with a marked hypercellularity (SLE) and in areas occupied by matrix nodules (diabetic nephropathy).

Abnormal glycosylation of serum IgA has been shown in IgA nephropathy. A significant down-regulation of VEGF mRNA and protein in human mesangial cells was developed after incubation with aberrantly glycosylated IgA68). Depressed VEGF synthesis may play a role in the aberrant vascular repair and favor sclerosis in IgA nephropathy.

Collectively, VEGF expression seems to be associated with the nature and pattern of glomerular diseases, and less so with the disease itself. VEGF expression was decreased in the regions with sclerosis and impaired capillary density. One possible explanation is that VEGF expression is decreased in chronic renal lesions. Though it is not certain whether VEGF expression in glomerular diseases is playing a causative role or just results from the disease process, it appears that the beneficial effect of VEGF supplementation may have pathophysiological ground.

3. Diabetic Nephropathy

Several studies show that high glucose increases the production of VEGF in a variety of cell types6971). The expression of VEGF mRNA and protein was enhanced after exposure to high glucose in both glomerular and tubular epithelial cells of the kidney44, 72). In addition, the up-regulation of VEFG and its receptors was demonstrated in the kidney of diabetic rats73, 74). In the study in OLETF rats described above, renal VEGF mRNA and glomerular VEGF immunoreactivity were reported to be increased over the duration of 9 to 68 weeks.

Besides the increased VEGF expression in hyperglycemia, there is evidence suggesting a potential pathophysiologic role of VEGF in DM nephropathy. High glucose induces VEGF gene and protein expression in several types of cells via protein kinase C, which is increasingly recognized as one of the central mediators of the damaging effects of hyperglycemia72, 75, 76). PKC inhibitor and PKC down-regulation inhibited glucose- induced increases in VEGF production70). In addition, a link between PKC and VEGF is further suggested by studies with an orally active PKCβ inhibitor77, 78). PKCβ inhibition was associated not only with reduced VEGF-induced retinal permeability but also with retardation of the development of albuminuria in streptozotocin-induced diabetic rats.

Another potentially important subject is the relation between VEGF and NO. VEGF exerts its angiogenic, vascular permeability and hemodynamic effects via activation and upregulation of eNOS in endothelial cells7981). The action of NO as a downstream mediator of VEGF is commensurate with the up-regulation of the NO system in early diabetes and is implicated in the pathogenesis of early renal dysfunction. NOS blockade substantially reduces or even completely eliminates the hyperfiltration in experimental diabetes8284). VEGF blockade prevented the up-regulation of eNOS in the diabetic glomeruli, thus supporting the contention that VEGF binds to its receptors on glomerular capillary endothelial cells and increases eNOS expression in these cells85). Along with this, interference with the VEGF-NO axis was shown to prevent microvascular dysfunction induced by high glucose level in experimental rats86).

The expression of VEGF in the diabetic kidney may depend not only on the effects of hyperglycemia but on the effects of advanced glycated end products (AGEs) that accumulate in diabetic tissues over weeks and months87). These AGEs have been shown to activate VEGF expression in vivo and in vitro. Various other factors relevant to the pathogenesis of diabetic nephropathy have also been shown to promote VEGF expression, including stretch88), angiotensin II33), and a number of cytokines27, 28). Recently, one study showed that anti-VEGF antibody improved early renal dysfunction such as, hyperfiltration, albuminuria and glomerular hypertrophy in diabetic rats85).

4. Renal Tumors

Since Folkman’s proposal in 1971 that inhibition of angiogenesis may be a valid strategy for the treatment of solid tumors89), extensive research has been dedicated to the identification and characterization of tumor angiogenesis factors. Many studies have demonstrated that VEGF is markedly up-regulated in the vast majority of human tumors so far examined, including lung, breast, gastrointestinal tract, bladder, ovary90).

In the kidney, VEGF has been known as a major tumor angiogenic factor. Increased serum levels of VEGF in patients with renal cell carcinoma (RCC) have been reported and these levels were also increased in the affected kidney of patients with RCC91). Furthermore, VEGF expression in RCC correlated positively with the grade and size of the tumor92, 93). Therefore, VEGF expression is a potentially significant independent predictor of the outcome. Besides the RCC, angiogenesis in Wilm’s tumor is also driven by VEGF94).

5. Other Diseases

The VEGF mRNA and protein expression are enhanced in the experimental chronic cyclosporine (CsA) nephrotoxicity model95). CsA also increases the expression of VEGF receptor mRNA. Since chronic low-grade tissue hypoxia is believed to be important in the development of chronic CsA nephrotoxicity96), CsA may cause injury to the endothelium, which may then up-regulate VEGF production as an adaptive mechanism directed toward repair and maintainance of the damaged endothelium. Actually, VEGF administration blunted the development of salt-sensitive hypertension and nephropathy after CsA exposure97).

VEGF protein expression was shown to be up-regulated in the kidney of patients with chronic renal allograft rejection, a renal disease with histologic similarities to chronic CsA nephrotoxicity98). Inhibition of VEGF by topically applied neutralizing antibody markedly suppresses acute rejection of rat corneal allografts, indicating a contributory role for VEGF in the pathogenesis of rejection99). Recently, one study showed that VEGF gene polymorphism is associated with acute renal allograft rejection and may be a useful marker of the risk for rejection100). It is suggested that increased VEGF production enhances endothelial permeability and augments leukocyte infiltration of the allograft, which may promote a clinically recognizable rejection episode. However, the exact mechanism (s) and role of VEGF in the allograft rejection remain to be determined.

One recent study discovered increased angiogenesis and VEGF expression in the renal cysts in patients with adult polycystic kidney disease (ADPKD)101). The epithelial cells of some cysts and enlarged tubules in more preserved areas express VEGF, and some capillaries surrounding the cysts express VEGF R-2. It can be suggested that cyst development is associated with the local enhancement of the microvascular network which, in turn, may facilitate or promote fluid secretion into the cysts. The postulated molecular mechanism of this process involves VEGF secretion and stimulation of neovascularization.

Pharmacological tools to modulate VEGF in the kidney

1. Suppressing effects of VEGF

First of all, many attempts using the angiogenic effect of VEGF have been made at anti-tumor therapy. There is an extensive body of evidence documenting the fact that inhibition of VEGF activity results in suppression of the growth of a wide variety of tumor cell lines in murine models102). Furthermore, clinical trials in cancer patients are under way with several VEGF inhibitors, including a humanized monoclonal antibody and various small molecules, inhibiting VEGF signal transduction102). In the kidney, some reports show that anti-VEGF antibody alone or in combination with other anti-angiogenic drugs is effective in suppressing the growth of Wilm’s tumor102, 104). Though not as well documented as the therapy of Wilm’s tumor, there is a report that anti-angiogenic agents inhibit the expression of VEGF105) in renal cell carcinoma. But, so far, there was no report about the direct relation between VEGF and renal cell carcinoma growth.

Based on the thought that an enhanced renal VEGF system is responsible for diabetes-associated renal changes, one report using anti-VEGF monoclonal neutralizing antibody in diabetic rats has recently been published85). Anti-VEGF antibody treatment decreased hyperfiltration, albuminuria and glomerular hypertrophy in streptozotocin-induced diabetes in rats. In an animal model of type II diabetes106), it was shown that long-term AGEs-inhibitor treatment abolished the enhanced renal VEGF expression and afforded a renoprotective effect by reducing diabetes-induced renal collagen IV accumulation to normal levels and reducing the albumin excretion rate.

So far, the use of anti-VEGF agents in ADPKD and renal allograft rejection has not been explored. However, since it is possible that VEGF is associated with the pathogenesis of ADPKD and chronic renal allograft rejection98, 101), anti-VEGF treatment has a potential for beneficial effects in those diseases.

2. Enhancing effects of VEGF

There have been numerous studies into the role of VEGF in renal failure. One study shows that VEGF blockade with an aptamer results in the inhibition of the capillary repair and leads to progressive renal damage107). In the model of experimental thrombotic microangiopathy, VEGF is capable of increasing peritubular capillary density, and this is associated with the amelioration of fibrosis and better preservation of renal function56, 108). Interestingly, while glomerular capillaries were repaired dramatically, peritubular capillary endothelium did not recover completely, and this was associated with a loss of VEGF from the tubular cells. VEGF can reduce the fibrosis and stabilize the renal function in the remnant kidney model109). In addition, in the CsA nephropathy, which is thought to be associated with chronic low-grade hypoxia, VEGF improves CsA-induced hypertension and nephropathy97).

Masuda et al. demonstrated that angiogenic capillary repair plays an important role in recovery from severe glomerular damage in rats with experimentally induced glomerulonephritis63). Systemic administration of VEGF after acute glomerular injury successfully induced glomerular repair and resolution of glomerulonephritis, associated with stimulation of angiogenesis and vascular remodeling. The use of VEGF as an angiogenic factor, therefore, could be potentially clinically useful for the treatment of glomerulonephritis accompanied by severe endothelial injury and capillary destruction.

References

1. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other diseases. Nat Med 1:27–31. 1995;
2. Ferrara N. Vascular endothelial growth factor. Eur J Cancer 32A:2413–2422. 1996;
3. Risau W. Mechanism of angiogenesis. Nature 386:671–674. 1997;
4. Alon T, Hemo I, Itin A, Pe’er J, Stone J, Keshet E. Vascular endothelial growth factor is a survival factor for newly-formed retinal vessels and has implications for retinopathy of prematurity. Nat Med 1:1024–1028. 1995;
5. Benjamin LE, Golijamin D, Itin A, Pode D, Keshet E. Selective ablation of immature blood vessels in established human tumors follows vascular endothelial growth factor withdrawal. J Clin invest 103:159–165. 1999;
6. Gerber HP, Dixit V, Ferrara N. VEGF induces expression of antiapoptotic proteins. J Biol Chem 273:13313–13316. 1998;
7. Gerber HP, McMurtrey A, Kowalski J, Yan M, Keyt BA, Dixit V, Ferrara N. VEGF regulates endothelial cell survival by the PI3-kinase/Akt signal transduction pathway. J Biol Chem 273:30336–30343. 1998;
8. Yuan F, Chen Y, Ellian M, Safabakhsh N, Ferrara N, Jain RK. Time-dependent vascular regression and permeability changes in established human tumor xenografts induced by an anti-vascular endothelial growth factor. Proc Natl Acad Sci USA 93:14765–14770. 1996;
9. Antonie DA, Barber AJ, Hollinger LA, Wolpert EB, Gardner TW. Vascular endothelial growth factor induces rapid phosphorylation of tight junction proteins, occluding and zonula occudin 1. J Biol Chem 274:23463–23467. 1999;
10. Tischer E, Mitchell R, Hartman T, Silva M, Gospodarowicz D, Fiddes JC, Abraham JA. The human gene for vascular endothelial growth factor. J Biol Chem 266:11947–11954. 1991;
11. Poltorak Z, Cohen T, Sivan R, Kandelis Y, Spira G, Vlodacsky I, Keshet E, Neufeld G. VEGF145, a secreted vascular endothelial growth factor isoform that binds to extra-cellular matrix. J Biol Chem 272:7151–7158. 1997;
12. Conn G, Bayne ML, Soderman DD, Kwok PW, Sullivan KA, Palisi TM, Hope DA, Thomas KA. Amino acid cDNA sequences of a vascular endothelial cell mitogen that is homologous to platelet-derived growth factor. Proc Natl Acad Sci USA 87:2628–2632. 1990;
13. Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor. Endocr Rev 18:4–25. 1997;
14. Ferrara N. Role of vascular endothelial growth factor in regulation of physiological angiogenesis. Am J Physiol Cell Physiol 280:C1358–C1366. 2001;
15. Park JE, Keller H-A, Ferrara N. The vascular endothelial growth factor isoforms(VEGF): differential deposition into the subepithelial extracellular matrix-bound VEGF. Mol Biol Cell 4:1317–1326. 1993;
16. Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor and its receptors. FASEB J 13:19–22. 1999;
17. Petrova TV, Makinen T, Alitalo K. Signaling via vascular endothelial growth factor receptors. Exp cell Res 253:117–130. 1999;
18. Achem MG, Jeltsch M, Kukk E, Makinen T, Vitali A, Wilks AF, Alitaio K, Stacker SA. Vascular endothelial growth factor D is a ligand for the tyrosine kinases VEGF receptor 2 and VEGF receptor 3. Proc Natl Acad Sci USA 95:779–791. 1997;
19. Joukov V, Pajusola K, Kaipainen A. A novel vascular endothelial growth factor, VEGF-C, is a ligand for the Flt-4 and KDR receptor tyrosine kinases. EMBO J 15:290–298. 1996;
20. Hideyasu O, Hitoshi T, Atsushi O, Shinji K, Seiji K, Akiyoshi U, Yoshihito H. Selective induction of neuropilin-1 by vascular endothelial growth factor. Proc Natl Acad Sci USA 99:383–388. 2002;
21. Minchenko A, Bauer T, Salceda S, Caro L. Hypoxic stimulation of vascular endothelial growth factor expression in vivo and in vitro. Lab Invest 71:374–379. 1994;
22. Shima DT, Adamis AP, Ferrara N, Yeo KT, Allende R, Folkman J, D’Amore PA. Hypoxic induction of endothelial cell growth factors in retinal cells. Mol Med 1:182–193. 1995;
23. Liu Y, Cox SR, Morita T, Kouremanas S. Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells. Circ Res 77:638–643. 1995;
24. Huang E, Arany Z, Livingston DM, Bunn HF. Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its submit. J Biol Chem 271:32253–32259. 1996;
25. Ikeda E, Achen MG, Breier G, Risau W. Hypoxia-induced trasncriptional activation and increased mRNA stability of VEGF in C6 glioma cells. J Biol Chem 270:19761–19766. 1995;
26. Levy AP, Levy NS, Goldberg MA. Post-transcriptional regulation of VEGF by hypoxia. J Biol Chem 271:2746–2753. 1996;
27. Frank S, Hubner G, Breier G, Longaker MT, Greenhalg DG, Werner S. Regulation of VEGF expression in cultured keratinocytes. J Biol Chem 270:12607–12613. 1996;
28. Cohen T, Nahari D, Cerem LW, Neufeld G, Levi BZ. Interleukin 6 induces the expression of VEGF. J Biol Chem 271:736–741. 1996;
29. Warren RS, Yuan H, Malti MR, Ferrara N, Donner DB. Induction of VEGF by insulin-like growth factor 1 in colorectal carcinoma. J Biol Chem 271:29483–29488. 1996;
30. Soh EY, Sobhi SA, Wong MG, Meng YG, Siperstein AE, Clark OH, Duh QY. TSH promotes the secretion of VEGF in thyroid cancer cell lines. Surgery 120:944–947. 1996;
31. Shifren JL, Mesiano S, Taylor RN, Ferrara N, Jaffe RB. Corticotropin regulates VEGF expression in human fetal adrenal cortical cells. J Clin Endocrinol Metab 83:1342–1347. 1998;
32. Amaral SL, Roman RJ, Greene AS. Renin gene transfer restores angiogenesis and VEGF expression in Dahl S rats. Hypertension 37:386–390. 2001;
33. Gruden G, Thomas S, Burt D, Ahou W, chusney G, Gnudi L, Viberti G. Interaction of angiotensin II and mechanical stretch on VEGF production by human mesangial cells. J Am Soc Nephrol 10:730–737. 1999;
34. Saijonmaa O, Nyman T, Kosonen R, Fyhrquist F. Up-regulation of angiotensin-converting enzyme by VEGF. Am J Physiol 280:H885–H891. 2001;
35. Hayashi K, Miyamoto A, Berisha B, Kosmann MR, Okuda K, Schams D. Regulation of angiotensin II production and angiotensin receptors in miscovascular endothelial cells from bovine corpus luteum. Biol Reprod 62:162–167. 2000;
36. Williams B, Baker A, Gallacher B, Ludwick D. Angiotensin II increases vascular permeability factor gene expression by human vascular smooth muscle cells. Hypertension 25:913–917. 1995;
37. Kang DH, Hughes J, Mazzali M, Schreiner GF, Johnson RJ. Impaired angiogenesis in the remnant kidney model (I): Potential role of vascular endothelial growth factor and thrombospondin-1. J Am Soc Nephrol 12:1434–1447. 2001;
38. Simon M, Grone H-J, Johren O, Kullmer J, Plate KH, Risau W, Fuchs E. Expression of VEGF and its receptors in human renal ontogenesis and in adult kidney. Am J Physiol 268:F240–F250. 1995;
39. Brown LF, Berse B, Tognazzi K, Manseau EJ, Van De Water L, Senger DR, Dvorak HF, Rosen S. Vascular permeability factor mRNA and protein expression in human kidney. Kidney Int 42:1457–1461. 1992;
40. Breier G, Albrecht U, Sterrer S, Risau W. Expression of VEGF during embryonic angiogenesis and endothelial cell differentiation. Development 114:521–532. 1992;
41. Marti H, Sisau W. Systemic hypoxia changes the organ-specific distribution of vascular endothelial growth factor and its receptors. Proc Natl Acad Sci USA 95:15809–15814. 1998;
42. Kanellis J, Mudge SJ, Fraser S, Katerelos M, Power DA. Redistribution of cytoplasmic VEGF to the bsaolateral aspect of renal tubular cells in ischemia-reperfusion injury. Kidney Int 57:2445–2456. 2000;
43. Awad BE, Kreft B, Wolber EM, Hellwig-birgel T, Metzen E, Fandrey J, Jelkmann W. Hypoxia and interleukin-1 β stimulate VEGF production in human proximal tubular cells. Kidney Int 58:43–50. 2000;
44. Kim BS, Chen J, Weinstein T, Noiri E, Goligorsky MS. VEGF expression in hypoxia and hyperglycemia: Reciprocal effect on branching angiogenesis in epithelial-endothelial co-cultures. J Am Soc Nephrol 13:2027–2036. 2002;
45. Witmer A, Dai J, Weich H, Vrensen G, Schingemann R. Expression of VEGF receptors 1,2, and 3 in quiescent endothelia. J Histochem Cytochem 50:767–777. 2002;
46. Thomas S, Vanuystel J, Gruden G, Rodriguez V, Burt D, Gnudi L, Hartley B, Viberti G. VEGF receptors in human mesangium in vitro and in glomerular disease. J Am Soc Nephrol 11:1236–1243. 2000;
47. Kanellis J, Fraser S, Katerelos M, Power DA. VEGF is a survival factor for renal tubular epithelial cells. Am J Phsiol 278:F905–F915. 2000;
48. Gengrinovitch S, Bermann B, David G, Witte L, Neufeld G, Ron D. Glypican-1 is a VEGF165 binding proteoglycan that acts as an extracellular chaperon for VEGF165. J Biol Chem 274:10816–10822. 1999;
49. Palade GE, Simionescu M, Simionescu N. Structural aspects of the permeability of the microvascular endothelium. Acta Physiol Scans Suppl 463:11–32. 1979;
50. Chen J, Braet P, Noiri E, Romanov V, Goligorsky MS. VEGF-induced mobilization of caveolae and permeability of endothelial cells. Am J Physiol: Cell 282:C1053–C1063. 2002;
51. LeCouter J, Kowalski J, Foster J, Hass O, Zhang Z, Dillard-Telm L, Franyz G, Rangell L, DeGuzman L, Keller GA, Peale F, Gurney A, Hillan KJ, Ferrara N. Identification of an angiogenic mitogen selective for endocrine gland epithelium. Nature 412:877–884. 2001;
52. Kendall RL, Thomas KA. Identification of a natural soluble form of the VEGF receptor FLT-1, and its heterodimerization with KDR. Biochem Biophys Res Commun 226:324–328. 1996;
53. Epstein FH, Agmon Y, Brezis M. Physiology of renal hypoxia. Ann NY Acd Sci 718:72–81. 1994;
54. Fine LG, Orphanides C, Norman JT. Progressive renal disease: the chronic hypoxia hypothesis. Kidney Int 65:S74–S78. 1998;
55. Norman JT, Clark IM, Garcia PL. Hypoxia promotes fibrogenesis in human renal fibroblasts. Kidney Int 58:2351–2366. 2000;
56. Kim YG, Suga S, Kang DH, Jefferson JA, Mazzali M, Gordon KL, Matsui K, Breiteneder-Geleff S, shankland SJ, Hughes J, Kerjaschki D, Schreiner G, Johnson RJ. VEGF accelerates renal a recovery in experimental thrombotic microangiopathy. Kidney Int 58:2390–2399. 2000;
57. Choi YJ, Chakraborty S, Nguyen V, Nguyen C, Kim BK, Shim SI, Suki WN, Truong LD. Peritubular capillary loss is associated with chronic tubulointersitial injury in human kidney: altered expression of VEGF. Hum Pathol 31:1491–1497. 2000;
58. Brenchley P. VEGF/VPF: A modulator of microvascular function with potential roles in glomerular pathophysiology. J Nephrol 9:10–17. 1996;
59. Noguchi K, Yoshikawa N, Ito-Kariya S, Inoue Y, Hayashi Y, Ito H, Nakamura H, lijima K. Activated mesangial cells produce vascular permeability factor in early-stage mesangial proliferative glomerulonephritis. J Am Soc Nephrol 9:1815–1825. 1998;
60. Heslan JM. The vascular permeability factor is a T lymphocyte a product. Nephron 42:187–188. 1986;
61. lijima K, Yoshikawa N, Connolly DT, Nakamura H. Human mesangial cells and peripheral blood mononuclear cells produce vascular permeability factor. Kidney Int 44:959–966. 1993;
62. Iruela-arispe L, Gordon K, Hugo C, Duijestijn AM, Claffey KP, Reilly M, Couser WG, Alpers CE, Johnson RJ. Participation of glomerular endothelial cells in the capillary repair of glomerulonephritis. Am J Pathol 147:1715–1727. 1995;
63. Masuda Y, Shimizu A, Mori T, ishiwata T, Kitamura H, Ohashi R, Ishizaki M, Asano G, Sugiyaki Y, Yamanaka N. VEGF enhances glomerular capillary repair and accelerates resolution of experimentally induced glomerulonephritis. Am J Pathol 159:599–608. 2001;
64. Webb NJA, Watson CJ, Roberts ISD, Bottomley MJ, Jones CA, Lewis MA, Postlethwaite RJ, Brenchley PEC. Circulating VEGF is not increased during relapses of steroid-sensitive nephrotic syndrome. Kidney Int 55:1063–1071. 1999;
65. Klanke B, Simon M, Rockl W, Weich HA, Stolte H, Grone HJ. Effects of VEGF/VPF on hemodynamics and permselectivity of the isolated perfused rat kidney. Nephrol Dial Transplant 13:875–885. 1998;
66. Honkanen E, Teppo AM, Gronhagen-Rlska C. Decreased urinary excretion of VEGF in idiopathic membranous glomerulonephritis. Kidney Int 57:2343–2349. 2000;
67. Shulman K, Rosen S, Tognazzi K, Manseau E, Brown A. Expression of vascular permeability factor is altered in many glomerular diseases. J Am Soc Nephrol 7:661–666. 1996;
68. Amore A, Conti G, Cirina P, Peruzzi L, Alpa M, Bussolino F, Coppo R. Aberrantly glycated IgA molecules down-regulate the synthesis and secretion of VEGF in human mesangial cells. Am J kidney Dis 36:1242–1252. 2000;
69. Natarajan R, Bai W, Lanting L, Ganzales N, Nadler J. Effects of high glucose on VEGF expression in vascular smooth muscle cells. Am J Physiol 273:H2224–H2231. 1997;
70. Cha DR, Kim NH, Yoon JW, Jo SK, Cho WY, Kim HK, Won NH. Role of VEGF in diabetic nephropathy. Kidney Int 58:S104–S112. 2000;
71. Sone H, Kawakami Y, Okuda Y, Kondo S, Hanatani M, Suzuki H, Yamashita K. VEGF is induced by long-term high glucose concentration and up-regulated by acute glucose deprivation in cultured bovine retinal pigmented epithelial cells. Biochem Biophys Res Commun 221:193–198. 1996;
72. Hoshi S, Nomoto K, Kuromotsu J, Tomari S, Nagata M. High glucose induced VEGF expression via PKC and ERK in glomerular podocytes. Biochem Biophys Res Commun 290:177–184. 2002;
73. Cooper ME, Vranes D, Youssef S, Stacker SA, Cox AJ, Rizkalla B, Casley DJ, Bach LA, Kelly DJ, Gilbert RE. Increased renal expression of VEGF and its receptor VEGF-2 in experimental diabetes. Diabetes 48:2229–2239. 1999;
74. Tsuchida K, Makita Z, Yamagishi S, Atsumi T, Miyoshi H, Obara S, Ishida M, Ishikawa S, Yasumura K, Koike T. Suppression of transforming growth factor beta and vascular endothelial growth factor in diabetic nephropathy in rats by a novel advanced glycation end product inhibitor, OPB-9195. Diabetologia 42:579–588. 1999;
75. Williams B, Gallacher B, Patel H, Orme C. Glucose-induced protein kinase C activation regulates vascular permeability factor mRNA expression and peptide production by humanvascular smooth muscle cells in vitro. Diabetes 46:1497–1503. 1997;
76. Uchida K, Uchida S, Nitta K, Yumura W, Marumo F, Nihei H. Glomerular endothelial cells in culture express and secrete VEGF. Am J Physiol 266:F81–F88. 1994;
77. Aiello LP, Bursell SE, Clermont A, Duh E, Ishii H, Takagi C, Mori F, Cillula TA, Ways K, Jirousek M, Smith LEH, King GL. VEGF-induced retinal permeability is mediated by protein kinase C in vivo and suppressed by an orally effective- βisoform-selective inhibitor. Diabetes 46:1473–1480. 1997;
78. Ishii H, Jisousek M, Koya D, Takagi C, Xia P, Clermont A, Bursell SE, Kern TS, Ballas LM, Heath WF, Stramm LE, King GL. Amelioration of vascular dysfunction in diabetic rats by an oral PKC beta inhibitor. Science 272:728–731. 1996;
79. Ziche M, Morbidelli L, Choudhuri R, Zhang HT, Donnini S, Granger HJ, Bicknell R. Nitric oxide synthase lies downstream from VEGF-induced but not basic fibroblastic growth factor-induced angiogenesis. J Clin Invest 99:2625–2634. 1997;
80. Tilton RG, Chang KC, Lejeune WS, Stephan CC, Brock TA, Williamson JR. Role for nitric oxide in the hyperpermeability and hemodynamic changes induced by intravenous VEGF. Invest Ophthalmol Vis Sci 40:689–696. 1999;
81. Hood JD, Meninger CJ, Ziche M, Granger HJ. VEGF up-regulates ecNOS message, protein and NO production in human endothelial cells. Am J Physiol 274:H1054–H1058. 1998;
82. Veelken R, Hilgers KF, Hartner A, Haas A, Bohmer KP, Sterzel RB. Nitric oxide synthase isoforms and glomerular hyperfiltration in early diabetic nephropathy. J Am Soc Nephrol 11:71–79. 2000;
83. Komers R, Allen TJ, Cooper ME. Role of endothelium-derived nitric oxide in the pathogenesis of renal hemodynamic changes of experimental diabetes. Diabetes 43:1190–1197. 1994;
84. Mattar AL, Fujihara CK, Ribeiro MO, de Nucci G, Zatz R. Renal effects of acute and chronic nitric oxide inhibition in experimental diabetes. Nephron 74:136–143. 1996;
85. Vriese AS, Tilton RG, Elger M, Stephan CC, Kriz W, Lameire NH. Antibodies against VEGF improve early renal dysfunction in experimental diabetes. J Am Soc Nephrol 12:993–1000. 2001;
86. Tilton RG, Kawamura T, Chang KC, Ido Y, Bjercke RJ, Stephan CC, Brock TA, Williamson JR. Vascular dysfunction induced by elevated glucose levels in rats is mediated by VEGF. J Clin Invest 99:2192–2202. 1997;
87. Bucala R, Vlassara H. Advanced glycation end products in diabetic renal and vascular disease. Am J Kidney Dis 26:875–888. 1995;
88. Gruden G, Thomas S, Burt D, Lane S, Chursney G, Sacks S, Viberti GC. Mechanical stretch induces vascular permeability factor in human mesangial cells. Proc Natl Acad Sci USA 94:12112–12116. 1997;
89. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 285:1182–1186. 1971;
90. Ferrara N. Molecular and biological properties of VEGF. J Mol Med 77:527–543. 1999;
91. Nicol D, Hii SI, Walsh M, The B, Thompson L, Kennett C, Gottey D. VEGF expression is increased in renal cell carcinoma. J Urol 157:1482–1486. 1997;
92. Tomisawa M, Tokunaga T, Oshika Y, Suto R, Yanagisawa K, Fukuda H, Nakano H, Abe S, Tateishi A, Kijimo H, Yamazaki H, Ueyama Y, Nakamura M. Expression pattern of VEGF isoform is closely correlated with tumor stage and vascularization in renal cell carcinoma. Eur J Cancer 35:133–137. 1999;
93. Paradis V, Lagha NB, Zeimoura L, Blanchet P, Eschwege P, Ba N, Benoit G, Jardin A, Bedossa P. Expression of VEGF in renal cell carcinoma. Virchows Arch 436:351–356. 2000;
94. Skoldenberg EG, Christiansson J, Sandstedt B, Larsson A, Lackgren G, Christofferson R. Angiogenesis and angiogenic growth factors in Wilm’s tumor. J urol 165:2274–2279. 2001;
95. Shihab FS, Bennett WM, Yi H, Andoh TF. Expression of VEGF and its receptors Flt-1 and KDR/Flk-1 in chronic cyclosporine nephrotoxicity. Transplant 72:164–168. 2001;
96. Bennett WM, Demattoes A, Meyer MM, Andoh T, Barry J. Chronic CsA nephropathy. Kidney Int 50:1089–1100. 1996;
97. Kang DH, Kim YG, Andoh TF, Gordon KL, Suga G, Mazzali M, Jefferson JA, Hughes J, Bennett W, Schreiner GF, Johnson RJ. Post-cyclosporine-demiated hypertension and nephropathy: amelioration by VEGF. Am J Physiol 280:F727–F736. 2001;
98. Pilmore HL, Eris JM, Painter DM, Bishop GA, McCaughan GW. VEGF expression in human chronic renal allograft rejection. Transplant 67:929–933. 1999;
99. Yatoh S, Kawakami Y, Imai M, Kozawa T, Segawa T, Suzuki H, Yamashita K. Effect of a topically applied neutralizing antibody against VEGF on corneal allograft rejection of rat. Transplant 66:1519–1524. 1998;
100. Shahbazi M, Fryer AA, Pravica V, Brogan IJ, Ramsay HM, Harden PN. VEGF gene polymorphism is associated with acute renal allograft rejection. J Am Soc Nephrol 13:260–264. 2002;
101. Bello-Reuss E, Holubec K, Rajarman S. Angiogenesis in autosomal-dominat polycystic kidney disease. Kidney Int 60:37–45. 2001;
102. Ferrara N. Role of VEGF in regulation of physiological angiogenesis. Am J Physiol 280:C1358–C1366. 2001;
103. Huang J, Moore J, Softer S, Kim E, Rowe D, Manley CA, O’Toole K, Middlersworth W, Stolar C, Yamashiro D, Kandel J. Highly specific antiangiogenic therapy is effective in suppressing growth of experimental Wilm’s tumors. J Pediatr Surg 36:357–361. 2001;
104. Softer S, Moore J, Kim E, Huang J, Manley CA, O’Toole K, Middlesworth W, Stolar C, Yamashiro D, Kandel J. Combination antiangiogenic therapy: increased efficacy in a murine model of Wilm’s tumor. J Pediatr Surg 36:1177–1181. 2001;
105. Sasamura H, Takahashi A, Miyao N, Maumori N, Kitamura H, Itoh N, Tsukamoto T. Inhibitory effect on expression of angiogenic factors by antiangiogenic agents in renal cell carcinoma. Br J Cancer 86:768–773. 2002;
106. Tsuchida K, Makita Z, Yamagishi S, Atsumi T, Miyoshi H, Obara S, Ishida M, Ishikawa S, Yasumura K, Koike T. Suppression of TGF-β and VEGF in diabetic nephropathy in rats by a novel advanced glycation end product inhibitor, OPB-9195. Diabetologia 42:579–588. 1999;
107. Ostendorf T, Kunter U, Loos A, Eitner F, Regele H, Kerjaschki D, Jangi N, Floege . VEGF165 mediates glomerular endothelial repair. J Clin Invest 104:913–923. 1999;
108. Suga S, Kim YG, Joly A, Puchartz E, Kang DH, Jefferson JA, Abraham JA, Hughes J, Johnson RJ, Schreiner GF. VEGF121 protects rats from renal infarction in a model of hemolytic uremic syndrome. Kidney Int 60:1297–1308. 2001;
109. Kang DH, Highes J, Mazzali M, Schreiner GF, Johnson RJ. Impaired angiogenesis in the remnant kidney model: II VEGF administration reduces renal fibrosis and stabilizes renal function. J Am Soc Nephrol 12:1448–1457. 2001;

Article information Continued

Figure 1.

Long-term effect of VEGF resulted in the formation of fenestration of endothelial cells. (A) HUVEC were cultured on matrigel for 36 h in the presence of 10 ng/mL VEGF-165. Light microscopic image shows an elaborate capillary-like network formed under these conditions. (B) Transmission electron micrograph shows diaphragmed fenestrae (arrowheads). (C) and (D) changes in electrical resistance and capacitance, respectively, in HUVEC treated with 10 ng/mL VEGF-165 (control cells were deprived of VEGF), demonstrating concomitant decrease in resistance and increase in capacitance compared with control. One-way ANOVA of experimental and control data in B and C showed that these curves are significantly different at p<0.05. HUVEC: human umbilical endothelial cell. Reprinted with permission from ref. 50.

Figure 2.

VEGF protein expression in GEC and RPTEC (A and B). Immunoprecipitation for VEGF in GEC (A) and RPTEC (B). Both bands shown are VEGF protein expressions from cell lysate and media, respectively. VEGF was detected using immunoprecipitation with polyclonal antibody followed by blotting with the monoclonal antibody. (C and D) VEGF expression in GEC (C) and RPTEC (D). GEC: rat glomerular epithelial cell, RPTEC: human renal proximal tubular epithelial cell; Con: control; D-glu: 30 mM D-glucose for 24 hours; L-glu: 30 mM L-glucose for 24 hours; Hypo: hypoxia for 24 hours; DG+H: 30 mM D-glucose+hypoxia for 24 hours.

*: p<0.01 compared with the control group, #: p<0.05 compared with the control group. Reprinted with permission from ref. 44.

Table 1.

Binding of VEGF family members to the known VEGF tyrosine kinase receptors

VEGF R1 (Flt-1) VEGFR2 (KDR/Flk-1) VEGF R3 (Flt-4)
VEGF-A Yes Yes No
VEGF-B Yes No No
VEGF-C No Yes Yes
VEGF-D No Yes Yes
VEGF-F No Yes No
PIGF Yes No No