Вы находитесь на странице: 1из 15

Kidney International, Vol. 65 (2004), pp.

2003–2017

PERSPECTIVES IN BASIC SCIENCE

The role of vascular endothelial growth factor (VEGF) in renal


pathophysiology
BIEKE F. SCHRIJVERS, ALLAN FLYVBJERG, and AN S. DE VRIESE
Renal Unit, Department of Internal Medicine, Gent University Hospital, Gent, Belgium; Medical Department M (Diabetes and
Endocrinology), Medical Research Laboratories, Institute of Experimental Clinical Research, Aarhus University Hospital, Aarhus,
Denmark; and Renal Unit, Department of Internal Medicine, AZ Sint-Jan AV, Brugge, Belgium

The role of vascular endothelial growth factor (VEGF) in renal Vascular endothelial growth factor (VEGF-A or
pathophysiology. Vascular endothelial growth factor (VEGF) VEGF), formerly called vasculotropin or vascular per-
is an endothelial-specific growth factor that promotes endothe- meability factor (VPF), belongs to a family of multipo-
lial cell proliferation, differentiation and survival, mediates
endothelium-dependent vasodilatation, induces microvascular tent cytokines, also including VEGF-B, -C, -D, -E, and
hyperpermeability and participates in interstitial matrix re- placenta growth factor [1]. Alternative exon splicing of a
modeling. In the kidney, VEGF expression is most promi- single VEGF gene results in at least six different isoforms.
nent in glomerular podocytes and in tubular epithelial cells, They are homodimeric glycoproteins of 121, 145, 165,
while VEGF receptors are mainly found on preglomerular, 183, 189, and 206 amino acids (VEGF 121−206 ) in humans
glomerular, and peritubular endothelial cells. The role of VEGF
in normal renal physiology is essentially unknown. The ab- and are one amino acid shorter in rodents [2]. VEGF 121 ,
sence of prominent effects of VEGF blockade in normal VEGF 165 , and VEGF 189 are the most abundantly ex-
experimental animals suggests a limited function during home- pressed isoforms, whereas VEGF 145 and VEGF 206 are
ostasis, although a role in the formation and maintenance of comparatively rare [2]. VEGF stimulates endothelial
glomerular capillary endothelial fenestrations has been sug- cell proliferation and differentiation, increases vascular
gested. VEGF and its receptors are up-regulated in experi-
mental animals and humans with type 1 and type 2 diabetes. permeability, mediates endothelium-dependent vasodi-
Inhibition of VEGF has beneficial effects on diabetes-induced latation, and supports vascular survival by preventing
functional and structural alterations, suggesting a deleterious endothelial apoptosis [1, 2]. In addition, VEGF induces
role for VEGF in the pathophysiology of diabetic nephropa- plasminogen activator, plasminogen activator inhibitor-
thy. VEGF is required for glomerular and tubular hypertro- 1 and interstitial collagenase, factors important in
phy and proliferation in response to nephron reduction, and
loss of VEGF is associated with the development of glomeru- matrix remodeling. Furthermore, VEGF promotes
losclerosis and tubulointerstitial fibrosis in the remnant kidney. monocyte chemotaxis and expression of adhesion
No firm conclusions on the role of VEGF in minimal change molecules [1, 2]. VEGF 165 , VEGF 189 , and VEGF 121 differ
or membranous glomerulonephritis can be drawn. VEGF may in affinity for heparin and heparan-sulfate proteoglycans
be an essential mediator of glomerular recovery in prolif- (VEGF 189 > VEGF 165 > VEGF 121 ) and in mitogenic ef-
erative glomerulonephritis. Glomerular and tubulointerstitial
repair in thrombotic microangiopathy and cyclosporin nephro- fect (VEGF 165 > VEGF 121 ) [2]. VEGF 165 , VEGF 189 , and
toxicity may also be VEGF-dependent. In conclusion, VEGF VEGF 206 are in most part sequestered in the extracellu-
is required for growth and proliferation of glomerular and per- lar matrix and at the cell surface, whereas VEGF 121 and
itubular endothelial cells. While deleterious in some, it may con- VEGF 145 are freely released [2]. The receptors for VEGF,
tribute to recovery in other forms of renal diseases. previously described as fms-like tyrosine kinase (Flt-1)
and fetal liver kinase 1 (Flk-1/KDR), now designated as
VEGFR-1 and VEGFR-2, respectively, are high-affinity
transmembrane tyrosine kinase receptors [2]. Soluble
VEGFR-1 (sVEGFR-1), a splice variant of VEGFR-1,
regulates VEGF availability by binding VEGF in the cir-
Key words: compensatory hypertrophy, diabetic nephropathy, glomeru-
lonephritis, transplant rejection, vascular endothelial growth factor
culation [3, 4]. Neuropilin-1 and Neuropilin-2 act as iso-
(VEGF), uremia. form specific co-receptors for VEGF [5]. Hypoxia is the
main stimulus for VEGF expression and/or production.
Received for publication April 24, 2003
and in revised form August 7, 2003, and September 25, 2003
Several growth factors and cytokines such as epidermal
Accepted for publication December 3, 2003 growth factor, transforming growth factor b (TGF-b),
platelet-derived growth factor (PDGF), insulin-like

C 2004 by the International Society of Nephrology growth factor I (IGF-I), angiotensin II, interleukin-1

2003
2004 Schrijvers et al: VEGF in renal pathophysiology

(IL-1), and IL-6 also have the potential to up-regulate ularly in podocytes [24]. VEGFR-1 and VEGFR-2 were
VEGF expression. VEGF may be induced by other fac- detected in cultured rat and human mesangial cells [25–
tors as well [i.e., prostaglandins, mechanical stress, hy- 28] and in cultured rat renal tubular epithelial cells [29],
perglycemia, advanced glycation end products (AGEs), but not in cultured primary human podocytes [30]. In con-
protein kinase C (PKC), and reactive oxygen species trast, conditionally immortalized human podocytes ex-
(ROS)]. VEGF up-regulates the expression of endothe- pressed VEGFR-1, VEGFR-3 and Neuropilin-1 but not
lial nitric oxide synthase (NOS3) in endothelial cells and VEGFR-2 [31]. Cultured mouse glomerular endothelial
increases the production of nitric oxide [6]. Several lines cells and transformed tubular epithelial cells expressed
of evidence have indicated that VEGF exerts its biologic Neuropilin-1 and Neuropilin-2 [32]. Neuropilin-1 was
effects through nitric oxide [7]. Nitric oxide may down- also detected in cultured human mesangial cells [25, 27]
regulate VEGF expression and thus function in a negative and in cultured primary human glomerular podocytes
feedback regulator mechanism [8]. Recently, 15 differ- [30]. The expression of VEGFR-1, VEGFR-2, sVEGFR-
ent sequence polymorphisms have been identified within 1, and Neuropilin-1 in isolated human glomeruli was
the VEGF gene, including a C/T base change at position also heterogenous [3, 30]. In human kidney, VEGFR-1
−460, a G/C change at +405 and a A/C change at −141 and VEGFR-2 were predominantly expressed on pre-
[9]. The −460C/+405G and −460T/+405C haplotypes are glomerular, glomerular, and peritubular endothelial cells
the most frequently observed in the normal population [20, 23, 27, 33]. In rat kidney, VEGFR-2 expression was
[9]. A correlation of the +405 genotype with production detected in glomerular and peritubular endothelial cells,
of VEGF has been demonstrated in vitro [9] and in vivo in distal convoluted tubules and collecting ducts, and
[10], with the highest VEGF production for the GG geno- in cortical interstitial fibroblast and medullary intersti-
type, intermediate production for the GC genotype and tial cells, whereas VEGFR-1 was expressed more diffuse
lowest production for the CC genotype [9]. Further, the in proximal and distal tubules [19, 29]. In human kid-
combination of the +405G genotype with other polymor- ney, Neuropilin-1 was detected in glomerular podocytes
phisms resulted in higher VEGF promotor activity [11]. [30]. Neuropilin-1 and Neuropilin-2 were localized in per-
A deletion/insertion (D/I) polymorphism at the −2549 itubular capillary endothelial cells in adult mouse and rat
position of the VEGF promotor region has been linked kidney [32]. Tie2 was demonstrated in glomerular cap-
to increased transcriptional activity [12]. illary endothelial cells of human and rat glomeruli and
Angiopoietins form another family of endothelial- in cultured human microvascular endothelial cells [24].
specific growth factors consisting of angiopoietin-1 In summary, in vivo, capillary endothelial cells express
(Ang-1) and angiopoietin-2 (Ang-2), which bind to tyro- VEGFR-1, VEGFR-2, and Tie2, glomerular podocytes
sine kinase receptors Tie1 and Tie2 [13]. Angiopoietins express Neuropilin-1 and produce VEGF and Ang-1.
and VEGF play co-ordinated and complementary roles Although the functions of constitutively expressed
in vascular homeostasis [13]. Ang-2 stimulates new blood VEGF and VEGF receptors in the normal kidney are
vessel formation in the presence of VEGF, but promotes largely unknown, some hypotheses may be derived from
endothelial apoptosis and vessel regression when VEGF the peculiar distribution of the molecule and its recep-
levels are low [14]. tors in the kidney. VEGF is strongly expressed by vis-
ceral epithelial cells while its binding sites are localized
on glomerular endothelial cells. If one assumes the ex-
VEGF IN RENAL PHYSIOLOGY istence of a paracrine loop in the glomerulus, VEGF
This section elaborates on the expression and the po- must move in the opposite direction of the glomerular
tential role of VEGF, angiopoietins, and their receptors filtrate in order to bind to its receptors. The presence
in the normal adult kidney. A comprehensive discussion of such complex mechanism suggests that the strategic
of the role of the VEGF system in renal development localization of podocytes is required for the correct sens-
is beyond the scope and space limitations of this re- ing and interpretation of the stimulus for VEGF release.
view and has been published elsewhere [15]. Cultured VEGF may be involved in the induction and maintenance
rat and human mesangial cells express both mRNA of of the fenestrae in glomerular capillary endothelial cells
VEGF 121 , VEGF 165 , and VEGF 189 , and VEGF protein [37]. Given the role of VEGF in promoting microvascu-
[16, 17]. In rodent and human kidneys, VEGF mRNA lar permeability, it has been speculated that VEGF may
and/or protein were detected predominantly in glomeru- regulate glomerular permeability, although it is generally
lar podocytes, distal tubules, and collecting ducts, and to acknowledged that the capillary fenestrations do not rep-
a lesser extent in some proximal tubules [3, 16, 18–23]. resent the ultimate barrier to filtration. Recently, in vitro
The expression of the different VEGF isoforms in normal evidence indicated that VEGF may act as an autocrine
human glomeruli was complex and variable with substan- factor on calcium homeostasis and cell survival in human
tial inter- and intraindividual variation [3]. Ang-1, but not podocytes [31]. In contrast to the prominent expression of
Ang-2, was identified in adult human glomeruli, partic- the VEGF system in the adult kidney, the administration
Schrijvers et al: VEGF in renal pathophysiology 2005

or inhibition of VEGF in normal adult animals appears to ing VEGF levels and parameters reflecting the sever-
have only minimal effects. In the isolated perfused rat kid- ity of diabetic nephropathy. VEGF levels were higher
ney, administration of VEGF increased the renal blood in macroalbuminuric type 1 diabetics than in patients
flow but did not influence the glomerular filtration rate or without complications [41, 45] and microalbuminuric pa-
the permselectivity of the glomerular barrier wall [34]. In tients [abstract; Abdel Aziz MY, Nephrol Dial Transplant
vivo infusions of VEGF into the renal artery of rats did 12:1538a, 1997]. Other studies reported no differences in
not influence protein excretion rate [34]. The administra- VEGF levels between type 1 diabetic patients with and
tion of neutralizing monoclonal anti-VEGF-antibodies to without (micro)albuminuria [43, 44, 46]. Correlations be-
normal rats had no effect on glomerular filtration rate or tween VEGF levels and glycemic control, severity of dia-
glomerular volume [35]. Injection of a VEGF 165 aptamer, betic nephropathy, and degree of UAE were observed in
an oligonucleotide-based VEGF 165 antagonist, in normal some [abstract; Abdel Aziz MY, Nephrol Dial Transplant
rats had no effect on kidney and glomerular morphology, 12:1538a, 1997] [41] but not in all studies [43, 44, 46, 47].
did not induce proteinuria and did not affect glomerular A positive correlation was found between serum VEGF
cell proliferation and the number of endothelial fenes- levels and nailfold capillary permeability in type 1 diabet-
trations [36]. In contrast, podocyte-specific heterozygous ics [47]. Urinary VEGF excretion was unchanged in pa-
and homozygous deletions of VEGF in mice resulted in tients with diabetic nephropathy compared with control
proteinuria and endotheliosis by 21/2 weeks of age, and subjects [48]. Renal biopsies from patients with diabetic
in perinatal lethality, respectively, with loss of endothelial nephropathy showed fewer glomerular VEGF mRNA
fenestrations or failure to form fenestrations [37]. Con- positive cells than biopsies from controls [18]. Glomeru-
versely, podocyte-specific overexpression of VEGF 165 led lar VEGF expression was highest in the patients with
to a collapsing glomerulopathy [37]. mildest sclerotic changes and was particularly strong in
viable glomerular podocytes [20], but reduced or absent
in sclerotic glomeruli [20, 49]. Atrophic tubules were neg-
VEGF IN RENAL PATHOPHYSIOLOGY
ative for VEGF mRNA, except for a weak signal in distal
Diabetic nephropathy tubules, whereas interstitial cells often had pronounced
Type 1 diabetes. VEGF mRNA and protein expres- VEGF mRNA and protein positivity [20]. In patients
sion were increased at the onset of diabetes in genetically with type 1 diabetes and a minimum diabetes duration
diabetic BioBreeding rats [38], in glomerular podocytes, of 10 years, the frequency of the +405 CC genotype,
distal tubules and collecting ducts after 3 weeks and 32 which is associated with lower VEGF protein produc-
weeks of streptozotocin (STZ)-diabetes in rats [19] and in tion in peripheral blood mononuclear cells (PBMC) [9],
renal tubular and vascular compartments in STZ-diabetic was increased compared with control subjects [abstract;
rats with superimposed hypertension [39]. More specifi- Summers AM, J Am Soc Nephrol 13:248A, 2002]. An in-
cally, the VEGF 164 and VEGF 188 isoforms increased af- crease in the −460 T allele was found in patients with mi-
ter STZ-diabetes induction which was reversed by insulin croalbuminuria or proteinuria compared to patients with
treatment [40]. Glomerular VEGFR-1 and VEGFR-2 normoalbuminuria [abstract; Summers AM, J Am Soc
mRNA expression were higher after 6 weeks of STZ- Nephrol 13:248A, 2002]. The D allele and DD genotype
diabetes [40]. Similarly, VEGFR-2 mRNA was increased of the VEGF D/I polymorphism at −2549 of the promo-
in glomerular and peritubular endothelial cells and inter- tor region were associated with susceptibility to diabetic
stitial cells after 3 weeks of STZ-diabetes but not after nephropathy in type 1 diabetics and the presence of the
32 weeks [19]. To assess the role of VEGF in the patho- D allele was linked to increased transcriptional activity
physiology of early renal dysfunction in diabetes, type 1 [12].
diabetic rats were treated with monoclonal neutralizing Type 2 diabetes. Renal and/or glomerular VEGF
anti-VEGF-antibodies for 6 weeks. Inhibition of VEGF mRNA were increased in diverse experimental mod-
abolished the diabetes-associated glomerular hyperfiltra- els of type 2 diabetes [i.e., modestly in insulin-resistant
tion, glomerular hypertrophy, and urinary albumin ex- Zucker fatty/fatty rats and more pronounced in Zucker
cretion (UAE) without an effect on metabolic control diabetic fatty (ZDF) rats [40], in db/db mice [50], and in
[35]. In addition, the diabetes-associated up-regulation Otsuka Long-Evans Tokushima fatty (OLETF) rats] [51].
of NOS3 expression was prevented, further supporting In OLETF rats, VEGF mRNA expression was increased
the evidence that nitric oxide acts as a downstream me- only in the early period of diabetic nephropathy [51]. In
diator of VEGF [35]. ZDF rats, VEGF mRNA levels rose early in the course of
Both increased [abstract; Abdel Aziz MY, Nephrol diabetes and remained elevated up to 7 months [52]. At
Dial Transplant 12:1538a, 1997] [41, 42] and unaltered 9 months, when glomerulosclerosis was most pro-
[43, 44] serum or plasma VEGF levels were observed nounced, renal VEGF mRNA levels were reduced [52].
in type 1 diabetic patients versus control subjects. Var- VEGFR-1 and VEGFR-2 mRNA were also elevated in
ious studies examined a correlation between circulat- ZDF rats [40]. Treatment with anti-VEGF-antibodies in
2006 Schrijvers et al: VEGF in renal pathophysiology

db/db mice attenuated the diabetes-associated increases Conclusion. Data regarding circulating VEGF levels
in kidney weight, glomerular volume and UAE, and in patients with diabetes are highly discrepant. As VEGF
abolished the increase in basement membrane thickness is a paracrine mediator, systemic levels may not ade-
and creatinine clearance [53]. In addition, the antibodies quately reflect changes in the local VEGF system [43]. In
tended to reduce the mesangial matrix expansion [53]. In addition, the assay methods vary substantially between
contrast, in Goto-Kakizaki rats, an experimental model of the studies (i.e., VEGF isoform specificity, detection of
lean type 2 diabetes, anti-VEGF-antibodies only tended total or free VEGF and interference with VEGF-binding
to reduce glomerular hypertrophy and had no effect on molecules such as a 2 -macroglobulin) [59]. Finally, deter-
UAE or creatinine clearance [abstract; Schrijvers BF, minations of serum and plasma VEGF levels may dif-
J Am Soc Nephrol 13:764A, 2002]. Treatment of OLETF fer, owing to release from platelets and leukocytes after
rats with temocapril or CS-866, an angiotensin II type 1 sampling [60, 61]. Renal expression of VEGF and its re-
receptor (AT1) antagonist, improved glomerulosclerosis, ceptors is more consistently up-regulated in experimental
reduced urinary protein excretion and VEGF staining, animals and patients with type 1 and type 2 diabetes, espe-
suggesting that AT1 may be involved in the overproduc- cially early in the course of diabetes (Table 1). Inhibition
tion of VEGF in diabetic nephropathy [54]. of VEGF resulted in beneficial effects on the diabetes-
Plasma VEGF levels were higher in type 2 diabetics associated renal changes, underlining a deleterious role
than in controls [55]. In another study, plasma VEGF for VEGF in the pathophysiology of diabetic nephropa-
levels were elevated only in type 2 diabetic patients thy. Although the cause of the up-regulation of the VEGF
with characteristics of atherosclerosis [56]. Plasma VEGF system in diabetes remains unknown, several factors rel-
tended to rise with increasing UAE [55], however, other evant to the pathogenesis of diabetic nephropathy have
studies reported no such correlation [47, 57]. In type 2 di- been shown to promote VEGF expression in different cell
abetic patients, VEGF did not correlate with serum cre- types, including hyperglycemia, AGEs, PKC, angiotensin
atinine [57] or capillary permeability [47]. There were II, various cytokines, and aldose reductase.
no differences in sVEGFR-1 plasma levels between dia-
betic patients with or without atherosclerosis and con-
trol subjects [56]. Urinary VEGF excretion increased High protein–induced nephropathy
with the progression of diabetic nephropathy and cor- One study examined the potential role of VEGF in
related weakly with the levels of serum creatinine, crea- high protein–induced renal and glomerular enlargement
tinine clearance, microalbuminuria, and proteinuria [16]. (Table 1) [62]. Mice fed a diet containing 45% protein
In biopsies with mild changes of diabetic nephropathy, exhibited early glomerular hypertrophy and kidney
VEGF was up-regulated in glomerular podocytes and enlargement compared with mice on a 20% protein con-
distal tubular cells. In biopsies with advanced changes, taining diet. Treatment with a monoclonal neutraliz-
VEGF staining was decreased or negative in sclerotic ing VEGF-antibody abolished the high protein–induced
glomeruli but remained intense in tubules [16]. More glomerular hypertrophy without affecting kidney or body
recently, in patients with type 2 diabetic nephropathy, weight. Kidney IGF-I was up-regulated in high protein–
glomerular VEGF mRNA expression, observed predom- fed mice and not affected by treatment with the VEGF-
inantly in podocytes, was higher than in normal kidneys, antibody. Consequently, as IGF-I has been implicated
but in contrast to the previous study, VEGF expression in high protein–induced renal growth, VEGF could be
was higher in patients with advanced renal morphologic a downstream mediator of IGF-I. This study illustrates
changes than in patients with mild-moderate mesangial that VEGF plays a major role in high protein-induced
expansion and tubulointerstitial changes [abstract; glomerular growth [62].
Kanesaki Y, J Am Soc Nephrol 13:8A, 2002]. In glomeruli
of type 2 diabetic patients, VEGF mRNA level was
inversely related to albumin excretion rate [abstract; Nephron reduction
Bortoloso E, Diabetologia 42:273A, 1999]. Tubuloint- Uninephrectomized mice are characterized by an in-
erstitial VEGF and VEGFR-1 mRNA were lower in creased glomerular volume and kidney weight in the rem-
patients with severe diabetic nephropathy [58]. While nant kidney and an early transient increase in kidney
the ratio between VEGF 121 and VEGF 165 correlated IGF-I concentration [63]. To explore a role for VEGF
with mesangial matrix expansion in one study [abstract; in compensatory renal changes after uninephrectomy,
Bortoloso E, Diabetologia 42:273A, 1999], no relation these animals were treated with anti-VEGF-antibodies,
with the severity of the histologic changes was found which abolished the glomerular enlargement and par-
in another [58]. As VEGF 121 is freely diffusable and tially blocked the renal growth without affecting re-
VEGF 165 is mostly bound to extracellular matrix, the nal IGF-I levels [63]. After a 75% surgical nephron
pathophysiologic relevance of the ratio between both iso- reduction, the remnant kidneys of C57Bl6xDBA2/F1
forms remains to be determined. mice displayed compensatory glomerular and tubular
Table 1. Summary of the expression and intervention data per disease type in experimental models and patient populations
Effects
Renal expression
Manipulation of GFR/creatine
Disease VEGF VEGFR-1 VEGFR-2 VEGF axis clearance/RF UAE KW/GV Others
Type 1 diabetes Exp ↑ (19,38–40) ↑ (40) ↑ (19,40) VEGF neutralizing ↓ (35) ↓ (35) ↓ (35) ↓ NOS3 (35)
antibodies (35)
Clin ↑ (20) ND ND ND
↓ (18,49)a
Type 2 diabetes Exp ↑ (40,50–52) ↑ (40) ↑ (40) VEGF neutralizing ↓ (53) ↔ (a) ↓ (53) ↔ (a) ↓ (53) ↔ (a) ↓ BMT (53)
antibodies (53, a)
↓ (52)a ACE inhibition, AT1 ↓ (54) ↓ VEGF, GS (54)
antagonism (54)
Clin ↑ (16, b) ↓ (58) ND ND
↓ (16)a
HPN Exp ND ND ND VEGF neutralizing ↔/↓ (62) ↔ Kidney IGF-I (62)
antibodies (62)
Clin ND ND ND ND
Nephron reduction Exp ↑ (64) ND ND VEGF neutralizing ↓ (63) ↔ Kidney IGF-I (63)
antibodies (63)
↓ (65–68) VEGF 121 (66) ↑ EC prolif, ↑ NOS3, ↓ F (66)
ACE inhibition (68) ↑ GFR (68) ↓ (68) ↔ (68) ↑ EC density, ↑ VEGF, ↓ GS (68)
Clin ND ND ND ND
MCN Exp ↓ (85) ↓ (85) ↓ (85) VEGF 165 antagonizing ↔ (36) ↔ Pc ultrastructure,
aptamer (36) ↔ EC fenestration (36)
↑ (86) ↑ (86) ↑ (86)
Clin ↑ (18) ND ND ND
↔ (49,90)
MGN Exp ND ND ND VEGF 165 antagonizing ↔ (36) ↔ Pc ultrastructure (36)
aptamer (36)
Clin ↔ (18,90) ND ND ND
↑ (49)b
↓ (49)a ↓ (94) T
MPGN Exp ↑ (27,101) ND ↑ (27,101) VEGF 165 antagonizing ↓ EC prolif, ↑ GS (36)
aptamer (36)
Schrijvers et al: VEGF in renal pathophysiology

↓ (103) + UNX VEGF 165 (104) ↑ EC prolif (104)


Clin ↑ (90)b ↑ (90) ↑ (90) ND
↓ (27,49,90)a
CGN Exp ↓ (105)a ND ND VEGF 165 (c) ↑ RF (c) ↓ (c) ↑ EC prolif (c)
Clin ↔ (49,88)b ND ND ND
↓ (49,88)a
FSGS Exp ↓ (d) ND ND ND
Clin ↔ (49,90)b ND ND ND
↓ (49,90)a
TMA/HUS Exp ↑ (106) ND ND VEGF 121 (107,108) ↑ RF (107) ↑ Capillary recovery (107),
preservation (108), ↓ F (107)
Clin ↓ (e) T ↑ (e) ↑ (e) ND
Acute rejection Exp ND ND ND ND
Clin ↔ (110) ND ND ND
Chronic rejection Exp ↔ (111) ND ND ND
Clin ↑ (20,110) ND ND ND
↓ (20)a
2007
2008 Schrijvers et al: VEGF in renal pathophysiology

hypertrophy with prominent cortical peritubular capil-


↑ Damage, BUN, hematuria

Abbreviations used: BMT, basement membrane thickness; CGN, crescentic glomerulonephritis; Clin, clinical; DM, diabetes mellitus; EC, endothelial cell; Exp, experimental; F, fibrosis; FSGS, focal segmental
glomerulosclerosis; GFR, glomerular filtration rate; GS, glomerulosclerosis; GV, glomerular volume; HPN, high protein–induced nephropathy; HUS, hemolytic uremic syndrome; IGF-I, insulin-like growth factor
I; KW, kidney weight; MCN, minimal change nephropathy; MGN, membranous glomerulonephritis; MPGN, mesangioproliferative glomerulonephritis; NOS3, endothelial nitric oxide synthase; Pc, podocyte; prolif,
proliferation; RF, renal function; T, tubuli; TMA, thrombotic microangiopathy; UAE, urinary albumin excretion; UNX, uninephrectomy; ↓ reduced; ↑ enhanced; ↔ no change; ND, no data; (a) abstract; Schrijvers BF,
J Am Soc Nephrol 13:764A, 2002; (b) abstract; Kanesaki Y, J Am Soc Nephrol 13: 8A, 2002; (c) abstract; Shimizu A, J Am Soc Nephrol 13:36A, 2002; (d) abstract; Kairaitis LK, J Am Soc Nephrol 13:336A, 2002; (e)
↓ VEGF, ↓ VEGFR-2, ↓ F (113)
lary growth, whereas another strain, FVB/N mice, addi-
↓ Structural damage (116) tionally developed severe tubulointerstitial lesions and
glomerulosclerosis [64]. While total kidney VEGF pro-
Others

tein levels were increased, VEGF immunostaining re-


vealed a decrease in proximal tubules, a marked increase
in some distal tubules and no change in the glomeruli. In
mice with severe tubulointerstitial injury, the changes
in the peritubular microcirculation and VEGF expres-
(115)

sion were more prominent [64]. In the rat, experimen-


tal nephron reduction resulted in early peritubular and
KW/GV

glomerular endothelial cell proliferation followed by


a progressive loss of peritubular and glomerular cap-
illaries, the latter associated with a decreased VEGF
staining in tubules and glomerular podocytes [65–68].
↓ (115)
UAE

In another study, VEGF mRNA and protein expression


Effects

was increased 2 weeks, but decreased 4 weeks after 5/6


nephrectomy [abstract; Horike H, J Am Soc Nephrol
13:164A, 2002]. Administration of VEGF 121 after ex-
GFR/creatine
clearance/RF

perimental nephron reduction increased glomerular and


peritubular endothelial cell proliferation and reduced
tubulointerstitial fibrosis [66]. In addition, VEGF 121 in-
creased renal NOS3 staining and 24-hour urinary nitrite
and nitrate excretion [66], which is consistent with the
observation that VEGF-induced angiogenesis is me-
Table 1. (Continued)

ACE inhibition, AT1


Manipulation of

antagonism (113)
VEGF neutralizing
antibodies (115)

diated by nitric oxide. NOS blockade with L-N(G)-


VEGF axis

VEGF 121 (116)

nitro-arginine-methyl-ester (L-NAME) in animals with


remnant kidneys accelerated the development of pro-
teinuria, glomerulosclerosis, and tubulointerstial fi-
brosis, along with more glomerular and peritubular
ND
ND
ND
ND
ND
ND
ND
ND
ND

endothelial cell loss and impaired endothelial pro-


liferation [67]. VEGF immunostaining in glomerular
Sclerotic glomeruli or crescents in crescentic glomerulonephritis; b Preserved glomeruli.
↑ (112–114)
VEGFR-2

podocytes and tubules was not different between the


↔ (121)
↑ (118)
↑ (120)

groups [67]. In rats with subtotal nephrectomy, treat-


ment with perindopril improved glomerular filtration rate
ND
ND

ND
ND

ND
ND

and endothelial cell density, reduced UAE and glomeru-


Renal expression

losclerosis and restored VEGF expression [68]. Taken


↑ (112–114)
VEGFR-1

together, the experimental studies (Table 1) suggest that


↔ (120)
↑ (118)

VEGF is required for the glomerular and tubular hyper-


ND
ND

ND
ND
ND
ND
ND

trophy and endothelial cell proliferation in response to


nephron reduction. Down-regulation of VEGF may re-
abstract; te Loo DM, J Am Soc Nephrol 13:251A, 2002.

sult in the development of glomerulosclerosis and tubu-


↑ (112–114)
VEGF

lointerstitial fibrosis.
↔ (119)

↔ (121)
↑ (117)
↑ (118)

↑ (123)
↓ (49)

Human data regarding uremia and VEGF expression


ND

ND
ND

ND

are rather scarce. VEGF plasma levels were higher in


uremic patients than in control subjects [55, 69]. The rea-
Clin

Clin

Clin

Clin

Clin
Exp

Exp

Exp

Exp

Exp

sons for higher VEGF levels in uremia are unknown, but


excess production, tissue hypoxia, or reduced clearance
of VEGF have been suggested [55, 68]. The negative
Ischemia/reperfusion

correlation between plasma VEGF levels and residual


nephrotoxicity

Lupus nephritis

renal function observed in peritoneal dialysis patients


Congenital NS
Cystic kidney
Cyclosporine

suggests that renal clearance contributes to the elimina-


diseases

tion of VEGF. Conversely, VEGF may have a negative


injury
Disease

impact on residual renal function [70].


a
Schrijvers et al: VEGF in renal pathophysiology 2009

Glomerulonephritis mRNA expression was up-regulated in podocytes in


Minimal change nephropathy. Minimal change MCN and correlated with the urinary protein excretion
nephropathy (MCN) has been speculated to result from [18]. In two patients with systemic lupus erythematosus
the release of soluble circulating factors by mononuclear (SLE) and minimal light microscopic changes VEGF ex-
cells that alter glomerular permeability. As VEGF may pression was normal [49]. The allele frequencies of the
be a regulator of glomerular permeability, the release of dinucleotide repeat polymorphisms within the VEGFR-
VEGF by PBMC has been scrutinized. In cultured PBMC 1 gene were not different between patients with MCN
from patients with MCN, the effects of various cytokines and normal controls [91]. Similarly, polymorphisms in the
on the secretion of VPF in the culture supernatant were VEGF gene promotor were not associated with the de-
investigated [71–80]. VPF is an older term for VEGF velopment of proteinuria in MCN [abstract; Watson CJ,
and is determined by quantifying the intradermal per- J Am Soc Nephrol 9:A2497, 1998]. Further, the genotype
meability in guinea pigs. Cultured PBMC, activated by frequencies of the VEGF gene polymorphisms −460 C/T,
concanavalin-A or lipopolysaccharide, secreted VPF [71– −141 A/C, and +405 G/C were not different in children
82] and the VPF levels in the culture supernatant were with SSNS versus controls [92].
higher in PBMC from nephrotic MCN patients than from The lack of consistent alterations in the VEGF system
patients in remission or control subjects [71–80]. IL-2, in experimental animals or patients with MCN (Table 1),
IL-12, IL-15, and IL-18 stimulated and TGF-b1, IL-4, IL- does not allow firm conclusions to be drawn about the
10, and IL-13 inhibited VPF release by activated PBMC role of VEGF in the pathophysiology of MCN. Method-
from patients with MCN [71–79]. Patients treated with ologic difficulties may interfere with the correct interpre-
steroids had lower VPF levels in PBMC supernatant than tation of the results. PBMC in culture may be in anaerobic
untreated patients [71–78]. In contrast, VEGF mRNA metabolism, which may affect the expression of VEGF.
expression in unstimulated PBMC was not different be- Pitfalls in the determination of circulating VEGF lev-
tween children with steroid-sensitive nephrotic syndrome els have been described above. Further, high urinary
(SSNS) in relapse and children with SSNS in remis- VEGF levels may merely reflect podocyte loss and uri-
sion [83] or controls [84]. The potential role of VEGF nary podocyte excretion [93] rather than an active in-
in the development of proteinuria was also investigated volvement of VEGF in the disease process. It has even
in experimental MCN [85, 86]. In puromycin aminonu- been suggested that urinary VEGF may be derived from
cleoside nephrosis (PAN) rats, an experimental model the circulation and as such may be nothing more than an
of human MCN, the renal mRNA expression of VEGF assay for proteinuria [89].
and its receptors VEGFR-2 and VEGFR-1 was down- Membranous glomerulonephritis. Cultured lympho-
regulated [85]. In contrast, in hyperalbuminemia-induced cytes from nephrotic membranous glomerulonephritis
rat nephrosis, another model of human MCN, VEGF and (MGN) subjects did not release VPF in their super-
its receptors were up-regulated and correlated with the natants [82]. In rats with passive Heymann nephritis, an
severity of proteinuria [86]. In PAN rats, treatment with experimental model of MGN, proteinuria, podocyte foot-
a VEGF 165 aptamer for 6 days did not affect proteinuria, process fusion and subepithelial immune deposits were
podocyte swelling, foot-process fusion, or glomerular en- not affected by treatment with a VEGF 165 aptamer for
dothelial fenestrations [36], suggesting that VEGF 165 is 7 days [36]. Urinary VEGF excretion was decreased in
not involved in the disease process, although a pathogenic patients with idiopathic MGN compared with control
role for other isoforms is not excluded. Plasma VEGF subjects, but did not correlate with serum VEGF, renal
concentrations (free VEGF 121 and VEGF 165 ) and function or proteinuria [48]. In the follow-up of some pa-
urine VEGF/creatinine ratios were not elevated during tients, however, a reduction of proteinuria was associated
relapses of childhood SSNS when compared with remis- with increasing urinary VEGF excretion and the change
sion and normal controls [84]. Likewise, plasma or uri- in proteinuria over one year correlated inversely with the
nary VEGF levels were not different between patients change in urinary VEGF [48]. As mentioned above, de-
with MCN in the nephrotic state and those in remis- creased VEGF excretion in MGN may be explained by
sion [83] or healthy controls [87, 88]. In contrast, urinary podocyte injury and increased VEGF excretion during
VEGF levels were increased in patients with MCN with clinical improvement by partial recovery of podocytes.
nephrotic syndrome when compared with patients with- In 10 nephrotic MGN patients with already a significant
out nephrotic syndrome and healthy controls, and corre- degree of glomerulosclerosis, there was no difference in
lated with the degree of proteinuria [89]. Urinary VEGF renal VEGF mRNA expression compared with normal
levels decreased rapidly after steroid therapy [89]. Pa- kidneys and no correlation between VEGF expression
tients with MCN in the nephrotic state and in remission and the degree of proteinuria [18]. Another study, in-
displayed a similar VEGF protein and mRNA expres- cluding two patients with membranous nephropathy, one
sion mainly localized in glomerular podocytes [90]. In idiopathic and one lupus nephritis class IV, reported kid-
contrast, in situ hybridization revealed that VEGF ney VEGF protein and mRNA expression to be similar
2010 Schrijvers et al: VEGF in renal pathophysiology

as in controls and localized mainly in podocytes [90]. In possibly due to impairment of glomerular endothelial
three other cases of MGN, VEGF mRNA and protein ex- repair [102, 103]. Rats with anti-Thy 1.1 nephritis com-
pression was strong in relatively preserved glomeruli but bined with uninephrectomy [103] or Habu-snake venom
decreased or absent in sclerotic areas [49]. A recent study injection [104] developed progressive glomerulosclerosis
demonstrated reduction of VEGF immunostaining in the and renal insufficiency. In the former model, regenera-
tubular epithelium of MGN patients versus controls [94]. tion of glomerular endothelial cells and VEGF mRNA
The C allele of the VEGF −460 C/T polymorphism was expression were decreased [103]. In the latter model,
associated with progression toward end-stage renal fail- the administration of recombinant human VEGF 165 en-
ure in patients with idiopathic membranous nephropa- hanced endothelial cell proliferation and glomerular
thy [abstract; Summers AM, J Am Soc Nephrol 13:260A, capillary repair [104]. Serum VEGF levels were not dif-
2002]. ferent in patients with IgAN compared with normal con-
No consistent pattern of changes in the VEGF system trols [87, 88]. In contrast, urinary VEGF levels were
emerges from these studies (Table 1). Difficulties with increased in patients with IgAN with nephrotic syndrome
the interpretation of urinary VEGF levels have been de- when compared with patients without nephrotic syn-
scribed above. The discrepant results of VEGF expres- drome and healthy controls, and correlated with the de-
sion in renal biopsies of patients with MGN are likely gree of proteinuria [89]. In patients with MPGN due to
due to differences in the degree of glomerulosclerosis, as IgAN, Henoch-Schönlein nephritis, non-IgA prolifera-
loss of podocytes implicates loss of VEGF. It thus remains tive glomerulonephritis and lupus nephritis, VEGF pro-
unclear whether VEGF plays a role in the pathophysiol- tein and mRNA were expressed by glomerular podocytes
ogy of MGN. and mesangial cells but were faint or absent in areas of
Membranoproliferative glomerulonephritis. The glomerulosclerosis (Table 1) [49, 90]. In patients with
POEMS syndrome is a multisystemic syndrome charac- early primary MPGN [90] or IgAN, expression of VEGF
terized by polyneuropathy, organomegaly, endocrinopa- and its receptors was up-regulated, but in IgAN patients
thy, M protein, and skin changes. Renal involvement can with more severe interstitial damage, VEGF staining
occur and has been described as membranoproliferative was decreased [abstract; Horike H, J Am Soc Nephrol
glomerulonephritis-like lesions [95]. Circulating VEGF 13:164A, 2002] [27]. In patients with MPGN, including
levels were elevated in POEMS syndrome compared IgAN, the presence of the VEGF −460 CC genotype or
with normal controls [96, 97] or with patients with pri- C allele was associated with progression toward end-stage
mary membranoproliferative glomerulonephritis [98]. renal disease [abstract; Summers AM, J Am Soc Nephrol
There was no difference in serum VEGF levels between 13:260A, 2002].
POEMS patients with and without renal involvement In conclusion, VEGF may be essential for glomerular
[98]. Further, serum VEGF levels in POEMS patients repair in MPGN. Depressed VEGF synthesis resulting
did not correlate with glomerular alterations [95]. from podocyte injury may contribute to endothelial cell
Mesangioproliferative glomerulonephritis (MPGN). loss and favor the development of glomerulosclerosis.
PBMC of patients with IgA nephropathy (IgAN) re- Crescentic glomerulonephritis. In mice with anti-
sponded similarly to cytokines as those of MCN pa- glomerular basement membrane glomerulonephritis, an
tients, with stimulation of VPF release by IL-2, IL-12, experimental model of crescentic glomerulonephritis,
IL-15, and IL-18 and inhibition by TGF-b1, IL-4, IL- loss of glomerular capillaries was temporally associated
10, and IL-13 [71–78]. In contrast, VEGF mRNA lev- with decreased VEGF, VEGFR-2, Ang-1, and Tie2 im-
els in unstimulated PBMC were not different between munostaining and up-regulation of Ang-2, especially in
IgAN patients and healthy volunteers and did not glomeruli with crescents or sclerosis (Table 1) [105]. In
correlate with clinical or pathologic parameters [99]. antiglomerular basement membrane glomerulonephritis
Incubation of human mesangial cells with aberrantly gly- rats, treatment with VEGF 165 resulted in recovery of the
cosylated IgA resulted in a down-regulation of VEGF necrotizing and crescentic lesions, proliferation of en-
mRNA expression and decreased VEGF release in dothelial cells and capillary repair, and improved renal
the culture medium [100]. During recovery of anti- function and proteinuria [abstract; Shimizu A, J Am Soc
Thy 1.1 nephritis, an experimental model of MPGN, Nephrol 13:36A, 2002]. Serum VEGF levels were higher
up-regulation of VEGF mRNA in podocytes and the in patients with crescentic glomerulonephritis, includ-
mesangial region, and of VEGFR-2 mRNA in the ing pauci-immune rapidly progressive glomerulonephri-
glomerular capillary walls was associated with endothe- tis, Henoch-Schönlein purpura nephritis and IgAN, than
lial cell proliferation (Table 1) [27, 101]. In accordance, in normal control subjects or in patients with MCN, IgAN,
administration of a VEGF 165 antagonizing aptamer re- and focal segmental glomerular sclerosis (FSGS) [49, 87,
duced glomerular endothelial cell proliferation and inhib- 88]. Serum VEGF levels correlated with crescent fre-
ited glomerular capillary repair resulting in glomerular quency, tubulointerstitial injury, and glomerular mono-
sclerosis [36]. Glucocortocoids decreased VEGF release cyte infiltration, but not with urinary protein excretion or
and aggravated proteinuria in anti-Thy 1.1 nephritic rats serum creatinine levels [49, 87, 88]. Serum VEGF levels
Schrijvers et al: VEGF in renal pathophysiology 2011

decreased after corticosteroid therapy [49, 88]. VEGF ex- glomerular and peritubular capillary endothelium [106].
pression was normal in glomeruli with preserved architec- An early transient increase in VEGF immunostain-
ture but decreased or absent in glomeruli compressed by ing was observed in glomerular podocytes and corti-
crescents and in sclerotic areas (Table 1) [49, 88]. Taken cal tubules, but despite proliferation of glomerular and
together, the experimental data suggest that VEGF peritubular endothelial cells, progressive glomerular and
accelerates glomerular recovery in crescentic glomeru- tubulointerstitial damage, and renal failure developed
lonephritis. Further, for the first time, a role for angiopoi- [106]. The hypothesis that angiogenic growth factors may
etins in glomerular disease is suggested. Up-regulation of accelerate recovery of renal microvascular injury was
Ang-2 may be an appropriate adaptive response to pro- studied in experimental TMA (Table 1) [107, 108]. Sub-
mote new vessel formation, but in the presence of low cutanous administration of VEGF 121 resulted in greater
VEGF levels it may contribute to endothelial apoptosis recovery of the renal microvasculature together with a
and vessel regression [14]. better renal function and less tubulointerstitial fibrosis
FSGS. VEGF mRNA expression in unstimulated [107]. VEGF 121 -treated rats had higher urinary nitrites
PBMC was higher in children with FSGS than in those and nitrates excretion, suggesting that the angiogenic ef-
with MCN but the difference was not significant [83]. fects of VEGF were mediated by nitric oxide [107]. In
Rats developed proteinuria after the injection of the su- a more severe form of renal TMA in rats with acute
pernatant of cultured PBMC from FSGS patients sug- massive renal infarction, VEGF administration resulted
gesting that a “glomerular permeability factor” released in reduced glomerular endothelial cell apoptosis, pre-
from PBMC may change glomerular permeability and served microvascular endothelium and less cortical and
result in proteinuria in FSGS [81]. In a murine FSGS medullary necrosis [108]. Elevated serum and plasma
model, adriamycin nephrosis, areas of interstitial expan- VEGF levels were found in HUS patients compared with
sion and tubular atrophy were associated with increased controls and correlated with the severity of the disease
staining of hypoxia inducable factor-1 (HIF-1) in tubular [abstract; te Loo DM, J Am Soc Nephrol 13:251A, 2002].
and interstitial cells, reduced VEGF staining and loss of In three patients with HUS, immunohistochemistry of
cortical microvasculature [abstract; Kairaitis LK, J Am renal biopsy material showed increased VEGFR-1 and
Soc Nephrol 13:336A, 2002). Plasma VEGF levels were VEGFR-2 in the glomeruli, and absent VEGF staining
higher in proteinuric patients with FSGS than in those in tubuli compared to controls [abstract; te Loo DM, J Am
with MCNS but the difference was not significant [83]. Soc Nephrol 13:251A, 2002). In conclusion, VEGF may
Serum VEGF levels in FSGS patients were not different be required for glomerular and tubulointerstitial repair
from those in control subjects [48, 87, 88]. Urinary VEGF in TMA.
excretion, on the other hand, was higher than normal
in patients with FSGS, but did not correlate with serum
VEGF, renal function, or proteinuria [48]. In patients Renal transplantation
with FSGS, VEGF protein and mRNA expression were Acute rejection. To study the possible relationships
normal in preserved glomeruli but absent or very low between VEGF gene polymorphisms and the risk of
in glomeruli with segmental or global sclerosis (Table 1) acute renal allograft rejection, single nucleotide substi-
[49, 90]. Rare polymorphisms in the VEGF gene promo- tutions in the VEGF gene were identified in 173 white
tor were not associated with the development of protein- renal allograft recipients [109]. The correlation between
uria in FSGS [abstract; Watson CJ, J Am Soc Nephrol VEGF gene polymorphisms and VEGF production was
9:A2497, 1998]. In contrast, the CC genotype and C al- investigated in vitro in PBMC from healthy individuals.
lele of the more common −460 C/T polymorphism were Homozygotes with −1154G/G genotype and −2578C/C
associated with progression to end-stage renal disease in genotype showed the greatest risk of rejection and had
patients with FSGS [abstract; Summers AM, J Am Soc the highest production of VEGF by stimulated PBMC
Nephrol 13:260A, 2002]. from healthy volunteers, as compared with −1154A/A
The low renal VEGF expression and increased urinary and −2578A/A genotypes, respectively. Heterozygotes
VEGF excretion in FSGS may be secondary to podocyte with −1154G/A and −2578C/A genotypes demonstrated
injury and loss in the urine. No solid conclusions on the an intermediate risk. In biopsies of patients with acute
role of VEGF in the pathophysiology of FSGS can thus rejection or temporary allograft dysfunction, VEGF im-
be drawn. munostaining appeared to be quite similar to that of nor-
mal human kidney, but biopsies from normal kidneys
were not included in this study (Table 1) [110].
Thrombotic microangiopathy (TMA)/hemolytic Chronic rejection. In the Fischer (F344) donor to
uremic syndrome (HUS) Lewis recipient rat renal allograft, a well-established ex-
A rat model of renal TMA with histologic features perimental model of chronic allograft rejection, no dif-
similar to human HUS was induced by antiglomerular ferences in VEGF mRNA expression were observed
endothelial cell antibodies which produced loss of between allografts and isografts at any time. VEGF
2012 Schrijvers et al: VEGF in renal pathophysiology

expression did not correlate with the extent of protein excretion was higher in cyclosporine-treated mice
macrophage or myofibroblast infiltration [111]. In kid- than in cyclosporine and VEGF-antibody–treated mice
neys from four patients with chronic vascular rejection, [115]. In vitro, the cyclosporine-induced toxicity in cul-
VEGF mRNA and protein expression was increased tured murine proximal tubular epithelial cells also in-
compared with normal kidneys, most notably in prox- creased in the presence of a VEGF antibody [115]. In
imal and distal tubular cells and interstitial cells and a rat model of chronic cyclosporine nephrotoxicity, ad-
to a lesser extent in vascular smooth muscle cells and ministration of exogenous VEGF 121 resulted in renopro-
the mononuclear inflammatory infiltrate [20]. Viable tective effects (i.e., osteopontin expression, macrophage
glomerular podocytes displayed marked VEGF mRNA infiltration, collagen III deposition were decreased versus
expression, but VEGF mRNA labeling was reduced control rats and afferent arteriolopathy was dramatically
or absent in sclerosed glomeruli [20]. Similarly, in- improved) [116]. Treatment with VEGF 121 also lowered
creased renal VEGF protein expression was observed blood pressure, which was suggested to be mediated by
in glomerular podocytes and mesangial cells, vascu- the accelerated recovery from tubulointerstitial and mi-
lar smooth muscle cells, and some endothelial cells crovascular injury in VEGF-treated rats [116]. The role
and tubulointerstitium of kidneys in 17 patients with of VEGF in human cyclosporine nephrotoxocity has not
chronic renal allograft rejection, particularly in intersti- been evaluated.
tial cells likely to be macrophages [110]. Renal VEGF In summary, the expression of the VEGF system is
immunostaining was increased in patients with chronic increased in experimental cyclosporine nephrotoxicity.
rejection when compared with patients with acute re- VEGF-blockade aggravated and VEGF-administration
jection or temporary allograft dysfunction [110]. The in- ameliorated the cyclosporine-induced injury, suggesting
creased VEGF expression in glomeruli and particularly a role for VEGF in the repair process induced by cy-
interstitium of human kidneys with chronic allograft re- closporine nephrotoxicity.
jection may be induced by hypoxia, as a consequence
of reduced blood flow [20]. Other hypotheses for the
up-regulation of VEGF include production of VEGF by Other kidney diseases
macrophages or proliferating glomerular mesangial cells Cystic kidney diseases. One experimental study inves-
[110]. Alternatively, VEGF may contribute to the recruit- tigated the expressions of VEGF, HIF-1a and HIF-3a
ment of macrophages into the interstitium. in polycystic kidney lesions that occurred spontaneously
Cyclosporine nephrotoxicity. In a rat model of chronic in two rats [117]. Compared with the kidneys of control
cyclosporine nephrotoxicity, VEGF mRNA and protein rats, VEGF protein expression was increased in the inner
expression as well as VEGFR-1 and VEGFR-2 mRNA stripe of the outer medulla where cystic alterations were
expression were up-regulated as early as 7 days after ex- prominent. HIF-1a and HIF-3a protein expressions were
posure (Table 1) [112–114]. The increased VEGF and increased in proximal tubuli and in the thin loop of Henle,
VEGFR-1 expression continued until day 28, whereas respectively. In kidneys from 14 patients with autosomal-
VEGFR-2 expression declined but remained higher than dominant polycystic kidney disease (ADPKD), increased
in control rats [112, 113]. Immunostaining for VEGF was angiogenesis was identified, especially in the cyst walls
particularly strong in proximal and distal tubular cells and and around the cysts [118]. In addition, VEGF protein
occasionally in glomerular podocytes [113]. Treatment was expressed in the epithelial cysts cells, VEGFR-2 pro-
with enalapril or losartan improved the tubulointerstitial tein in some capillaries surrounding the cysts and some
fibrosis and afferent arteriolopathy, and decreased VEGF glomeruli, whereas VEGFR-1 was expressed irregularly
mRNA and protein expression and VEGFR-2 mRNA ex- in some cyst cells and remnant tubular cells. Furthermore,
pression [113]. L-NAME worsened both glomerular fil- VEGF 165 was demonstrated in cultured ADPKD cells
tration rate and cyclosporine-induced interstitial fibrosis and in their supernatant, and VEGF secretion was in-
and arteriolopathy, and further increased VEGF mRNA creased during hypoxia [118]. In patients with acquired
and protein expression while L-arginine had the oppo- cystic kidney disease, IL-6, IL-8, and VEGF concentra-
site effect, suggesting that nitric oxide down-regulates tions were higher in the cyst fluid than in the blood, and
VEGF expression in cyclosporine nephrotoxicity [114]. higher than in patients with other cystic nephropathies
In a mouse model of acute cyclosporine toxicity, spe- (i.e., ADPKD or renal cell carcinoma). Taken together,
cific blockade of VEGF by a monoclonal antibody in- these findings suggest an angiogenic role for VEGF in the
creased the deleterious effects of cyclosporine on the kid- pathogenesis of cystic kidney diseases, likely triggered by
ney [115]. Mice treated with the VEGF-antibody showed local hypoxia [117] and involving the development of a
enhanced tubular toxicity, more apoptotic nuclei, in- pericystic circulation which may be necessary for cyst cells
creased blood urea nitrogen and hematuria compared to grow [118].
with mice treated with cyclosporine alone or with cy- Ischemia/reperfusion injury. Cultured rat kidney
closporine and an irrelevant IgG [115]. However, urinary tubular epithelial cells subjected to hypoxia showed
Schrijvers et al: VEGF in renal pathophysiology 2013

increased VEGF staining in the periphery of the cells and an anti-VEGF pegylated aptamer (EYE001) are cur-
[119]. In a rat model of renal ischemia/reperfusion injury, rently evaluated in clinical trials to treat various types
whole kidney VEGF mRNA and protein expression was of cancer, as well as macular degeneration and dia-
not increased following ischemia or ischemia and reper- betic retinopathy [127, 128]. VEGF receptor antagonists
fusion, although preexisting VEGF in tubular epithelial are also available. SU5416 and ZM323881 are specific
cells redistributed from the cytoplasm to the basolateral for VEGFR-2, whereas PTK787/ZK222584 inhibits both
surface [119]. Further, VEGFR-1 mRNA expression was VEGFR-1 and VEGFR-2 [126]. Other strategies to re-
not changed, whereas VEGFR-2 mRNA expression was duce VEGF-mediated effects, including placenta growth
up-regulated most prominent in glomerular endothelial factor inhibition and drugs that interfere with signaling
cells and peritubular capillaries, but also in some tubu- molecules in the VEGF signal transduction pathway such
lar epithelial cells [120]. It was speculated that the in- as Src/Fyn kinase inhibitors, are in development or in
creased VEGFR-2 expression in peritubular capillaries early stages of testing [126]. Targeting specific VEGF sig-
during ischemia/reperfusion injury may direct the effects naling molecules might be a way to distinctively inhibit
of VEGF released by ischemic tubular epithelial cells to specific actions of VEGF.
adjacent endothelial cells in order to preserve the cap- Interference with the renin-angiotensin system may be
illary blood supply and to promote tubular cell survival an indirect means to affect the VEGF axis, but the in-
and recovery [120]. teraction between both systems is complex. In cultured
Congenital nephrotic syndrome of the Finnish type. In mesangial cells angiotensin II induced VEGF expression
kidney samples of infants with congenital nephrotic syn- [129], whereas in tubular epithelial cells angiotensin II
drome of the Finnish type, expression and localization of diminished VEGF expression [65]. In agreement, diver-
VEGF and VEGFR-2 mRNA was similar to normal adult gent effects have been reported with in vivo blockade
human kidney [121]. VEGF protein expression was also of the renin-angiotensin system. Angiotensin-converting
not different from normal kidneys except for an intense enzyme (ACE) inhibition or AT1 antagonism reduced
juxtaglomerular staining [121]. VEGF expression in diabetic nephropathy [54] and in
Lupus nephritis. VEGF mRNA in PBMC of patients cyclosporine nephrotoxicity [113], suggesting that AT1
with SLE did not differ from those of controls [122]. may be involved in the VEGF overexpression observed
VEGF plasma levels were higher in SLE patients than in in these pathologies. In contrast, ACE inhibition was as-
controls, and SLE patients with renal failure had higher sociated with an increased VEGF expression in the rem-
levels than those with normal renal function [123]. VEGF nant kidney [68].
protein expression was increased in distal tubules, col-
lecting ducts and some podocytes in SLE patients with
moderate renal failure [123]. In two cases of SLE with CONCLUSION
diffuse endocapillary proliferative glomerulonephritis, The strategic localization of VEGF in the vicinity of the
renal VEGF expression was reduced [49]. filtration barrier and its known effects on microvascular
Wegener’s granulomatosis. Serum VEGF levels were permeability have engendered the hypothesis that VEGF
higher in patients with Wegener’s granulomatosis than controls glomerular permeability in the normal adult kid-
in normal controls [124]. Serum VEGF levels were ney and induces proteinuria in pathologic conditions. Al-
markedly elevated in patients with major disease versus though a large number of studies have been designed to
minor disease activity, suggesting that VEGF may be a examine this hypothesis, none has been able to firmly
marker of disease activity [124]. support it. Correlations of plasma or urinary VEGF lev-
Renal cell carcinoma. VEGF promotes the growth els with proteinuria in diverse glomerular pathologies
of renal cell carcinoma, by virtue of its angiogenesis- have been inconsistent. Positive correlations between uri-
inducing potential [125]. A detailed description of the nary VEGF levels and proteinuria may relate to urinary
role of VEGF in renal cell carcinoma is, however, beyond podocyte loss rather than to a causative link between
the scope and space limitations of this review. renal up-regulation of VEGF and development of
proteinuria. The inhibition of VEGF in experimental
glomerulonephritis did not affect proteinuria. The laud-
INTERFERENCE WITH THE VEGF AXIS able effect of VEGF-blockade on proteinuria in experi-
Several strategies exist to target VEGF and its re- mental diabetes may be indirect through inhibition of the
ceptors, including VEGF neutralizing antibodies, VEGF disease process. Finally, podocyte-specific overexpression
antagonizing aptamers, VEGF receptor-blocking anti- of VEGF caused a collapsing glomerulopathy rather than
bodies, VEGF receptor antagonists, and angiopoietins proteinuria.
[126]. So far, the experience with these treatments in The paramount task of VEGF in the adult glomerulus
renal disease is limited to animal models. A human- appears to be the stimulation of capillary endothelial cell
ized anti-VEGF monoclonal antibody (bevacizumab) growth and proliferation. This may be inappropriate in
2014 Schrijvers et al: VEGF in renal pathophysiology

diabetic nephropathy where it contributes to glomerular 11. STEVENS A, SODEN J, BRENCHLEY PE, et al: Haplotype analysis of
hypertrophy and hyperfiltration, but may be an essential the polymorphic human vascular endothelial growth factor gene
promoter. Cancer Res 63:812–816, 2003
repair mechanism in glomerulonephritis and TMA. Sim- 12. YANG B, CROSS DF, OLLERENSHAW M, et al: Polymorphisms of the
ilarly, tubular cells may respond to hypoxia or injury with vascular endothelial growth factor and susceptibility to diabetic
the production of VEGF that stimulates proliferation of microvascular complications in patients with type 1 diabetes mel-
litus. J Diabetes Complications 17:1–6, 2003
peritubular capillaries in order to overcome the tubular 13. SATCHELL SC, MATHIESON PW: Angiopoietins: Microvascular mod-
damage. ulators with potential roles in glomerular pathophysiology. J
As the VEGF system is affected in a wide variety of kid- Nephrol 16:168–178, 2003
14. LOBOV IB, BROOKS PC, LANG RA: Angiopoietin-2 displays VEGF-
ney diseases, interventions to manipulate VEGF may be dependent modulation of capillary structure and endothelial cell
promising therapeutic tools. Several strategies to either survival in vivo. Proc Natl Acad Sci USA 99:11205–11210, 2002
inhibit or enhance the VEGF axis have shown promising 15. ROBERT B, ABRAHAMSON DR: Control of glomerular capillary
development by growth factor/receptor kinases. Pediatr Nephrol
results in animal models of renal disease, but no data in 16:294–301, 2001
humans are presently available. 16. CHA DR, KIM NH, YOON JW, et al: Role of vascular endothe-
The VEGF gene is highly polymorphic and certain lial growth factor in diabetic nephropathy. Kidney Int 58 (Suppl
77):S104–S112, 2000
polymorphisms may be associated with alterations in 17. IIJIMA K, YOSHIKAWA N, CONNOLLY DT, NAKAMURA H: Human
the expression of VEGF. Although the diagnostic im- mesangial cells and peripheral blood mononuclear cells produce
portance of genotyping renal patients remains to be vascular permeability factor. Kidney Int 44:959–966, 1993
18. BAILEY E, BOTTOMLEY MJ, WESTWELL S, et al: Vascular endothelial
established, some VEGF polymorphisms may develop growth factor mRNA expression in minimal change, membranous,
into useful markers of disease susceptibility and/or and diabetic nephropathy demonstrated by non-isotopic in situ
progression. hybridisation. J Clin Pathol 52:735–738, 1999
19. COOPER ME, VRANES D, YOUSSEF S, et al: Increased renal expres-
sion of vascular endothelial growth factor (VEGF) and its receptor
ACKNOWLEDGMENTS VEGFR-2 in experimental diabetes. Diabetes 48:2229–2239, 1999
20. GRÖNE HJ, SIMON M, GRÖNE EF: Expression of vascular endothe-
B. Schrijvers is supported by the Institute for the Promotion of lial growth factor in renal vascular disease and renal allografts. J
Innovation by Science and Technology in Flanders (IWT), A. Flyvb- Pathol 177:259–267, 1995
jerg is supported by the Danish Diabetes Association, the Eva and 21. KANG DH, ANDERSON S, KIM YG, et al: Impaired angiogene-
Henry Frænkels Memorial Foundation, and the Danish Medical Re- sis in the aging kidney: Vascular endothelial growth factor and
search Council. thrombospondin-1 in renal disease. Am J Kidney Dis 37:601–611,
2001
Reprint requests to An De Vriese, Renal Unit, AZ Sint-Jan AV Rud- 22. KRETZLER M, SCHRÖPPEL B, MERKLE M, et al: Detection of mul-
dershove 10, B-8000 Brugge, Belgium. tiple vascular endothelial growth factor splice isoforms in single
E-mail: an.devriese@UGent.be glomerular podocytes. Kidney Int 41 (Suppl 67):S159–S161, 1998
23. SIMON M, GRÖNE HJ, JOHREN O, et al: Expression of vascular en-
REFERENCES dothelial growth factor and its receptors in human renal ontogen-
esis and in adult kidney. Am J Physiol 268:F240–F250, 1995
1. FERRARA N, GERBER HP: The role of vascular endothelial growth 24. SATCHELL SC, HARPER SJ, TOOKE JE, et al: Human podocytes ex-
factor in angiogenesis. Acta Haematol 106:148–156, 2001 press angiopoietin 1, a potential regulator of vascular endothelial
2. NEUFELD G, COHEN T, GENGRINOVITCH S, POLTORAK Z: Vascular growth factor. J Am Soc Nephrol 13:544–550, 2002
endothelial growth factor (VEGF) and its receptors. FASEB J 25. AMEMIYA T, SASAMURA H, MIFUNE M, et al: Vascular endothelial
13:9–22, 1999 growth factor activates MAP kinase and enhances collagen syn-
3. WHITTLE C, GILLESPIE K, HARRISON R, et al: Heterogeneous vascu- thesis in human mesangial cells. Kidney Int 56:2055–2063, 1999
lar endothelial growth factor (VEGF) isoform mRNA and recep- 26. TAKAHASHI T, SHIRASAWA T, MIYAKE K, et al: Protein tyrosine ki-
tor mRNA expression in human glomeruli, and the identification nases expressed in glomeruli and cultured glomerular cells: Flt-1
of VEGF148 mRNA, a novel truncated splice variant. Clin Sci and VEGF expression in renal mesangial cells. Biochem Biophys
97:303–312, 1999 Res Commun 209:218–226, 1995
4. HORNIG C, BARLEON B, AHMAD S, et al: Release and complex for- 27. THOMAS S, VANUYSTEL J, GRUDEN G, et al: Vascular endothelial
mation of soluble VEGFR-1 from endothelial cells and biologial growth factor receptors in human mesangium in vitro and in
fluids. Lab Invest 80:443–54, 2000 glomerular disease. J Am Soc Nephrol 11:1236–1243, 2000
5. NAKAMURA F, GOSHIMA Y: Structural and functional relation of 28. TRACHTMAN H, FUTTERWEIT S, FRANKI N, SINGHAL PC: Effect of vas-
neuropilins. Adv Exp Med Biol 515:55–69, 2002 cular endothelial growth factor on nitric oxide production by cul-
6. HOOD JD, MEININGER CJ, ZICHE M, GRANGER HJ: VEGF upregu- tured rat mesangial cells. Biochem Biophys Res Commun 245:443–
lates ecNOS message, protein, and NO production in human en- 446, 1998
dothelial cells. Am J Physiol 274:H1054–H1058, 1998 29. KANELLIS J, FRASER S, KATERELOS M, POWER DA: Vascular en-
7. TILTON RG, CHANG KC, LEJEUNE WS, et al: Role for nitric oxide dothelial growth factor is a survival factor for renal tubular epithe-
in the hyperpermeability and hemodynamic changes induced by lial cells. Am J Physiol Renal Physiol 278:F905–F915, 2000
intravenous VEGF. Invest Ophthalmol Vis Sci 40:689–696, 1999 30. HARPER SJ, XING CY, WHITTLE C, et al: Expression of neuropilin-1
8. TSURUMI Y, MUROHARA T, KRASINSKI K, et al: Reciprocal relation by human glomerular epithelial cells in vitro and in vivo. Clin Sci
between VEGF and NO in the regulation of endothelial integrity. 101:439–446, 2001
Nat Med 3:879–886, 1997 31. FOSTER RR, HOLE R, ANDERSON K, et al: Functional evidence that
9. WATSON CJ, WEBB NJ, BOTTOMLEY MJ, BRENCHLEY PE: Identifi- vascular endothelial growth factor may act as an autocrine factor
cation of polymorphisms within the vascular endothelial growth on human podocytes. Am J Physiol Renal Phsysiol 284:F1263–
factor (VEGF) gene: Correlation with variation in VEGF protein F1273, 2003
production. Cytokine 12:1232–1235, 2000 32. VILLEGAS G, TUFRO A: Ontogeny of semaphorins 3A and 3F and
10. AWATA T, INOUE K, KURIHARA S, et al: A common polymorphism in their receptors neuropilins 1 and 2 in the kidney. Gene Expr Pat-
the 5 -untranslated region of the VEGF gene is associated with di- terns 2:151–155, 2002
abetic retinopathy in type 2 diabetes. Diabetes 51:1635–1639, 2002 33. SIMON M, RÖCKL W, HORNIG C, et al: Receptors of vascular
Schrijvers et al: VEGF in renal pathophysiology 2015

endothelial growth factor/vascular permeability factor 53. FLYVBJERG A, DAGNÆS-HANSEN F, DE VRIESE AS, et al: Ameliora-
(VEGF/VPF) in fetal and adult human kidney: Localization tion of long-term renal changes in obese type 2 diabetic mice by a
and [125I] VEGF binding sites. J Am Soc Nephrol 9:1032–1044, neutralizing vascular endothelial growth factor antibody. Diabetes
1998 51:3090–3094, 2002
34. KLANKE B, SIMON M, RÖCKL W, et al: Effects of vascular endothe- 54. KOGA K, YAMAGISHI S, TAKEUCHI M, et al: CS-886, a new angiotensin
lial growth factor (VEGF)/vascular permeability factor (VPF) on II type 1 receptor antagonist, ameliorates glomerular anionic site
haemodynamics and permselectivity of the isolated perfused rat loss and prevents progression of diabetic nephropathy in Otsuka
kidney. Nephrol Dial Transplant 13:875–885, 1998 Long-Evans Tokushima fatty rats. Mol Med 8:591–599, 2002
35. DE VRIESE AS, TILTON RG, ELGER M, et al: Antibodies against 55. WASADA T, KAWAHARA R, KATSUMORI K, et al: Plasma concentra-
vascular endothelial growth factor improve early renal dysfunction tion of immunoreactive vascular endothelial growth factor and its
in experimental diabetes. J Am Soc Nephrol 12:993–1000, 2001 relation to smoking. Metabolism 47:27–30, 1998
36. OSTENDORF T, KUNTER U, EITNER F, et al: VEGF(165) mediates 56. BLANN AD, BELGORE FM, MCCOLLUM CN, et al: Vascular endothe-
glomerular endothelial repair. J Clin Invest 104:913–923, 1999 lial growth factor and its receptor, Flt-1, in the plasma of patients
37. EREMINA V, SOOD M, HAIGH J, et al: Glomerular-specific alterations with coronary or peripheral atherosclerosis, or type II diabetes.
of VEGF-A expression lead to distinct congenital and acquired Clin Sci 102:187–194, 2002
renal diseases. J Clin Invest 111:707, 2003 57. SHIMADA K, BABA T, NEUGEBAUER S, et al: Plasma vascular en-
38. BRAUN L, KARDON T, REISZ-PORSZASZ ZS, et al: The regulation of dothelial growth factor in Japanese type 2 diabetic patients with
the induction of vascular endothelial growth factor at the onset and without nephropathy. J Diabetes Complications 16:386–390,
of diabetes in spontaneously diabetic rats. Life Sci 69:2533–2542, 2002
2001 58. BORTOLOSO E, DEL PRETE D, GAMBARO G, et al: Vascular en-
39. CHENG HF, WANG CJ, MOECKEL GW, et al: Cyclooxygenase-2 in- dothelial growth factor (VEGF) and VEGF receptors in diabetic
hibitor blocks expression of mediators of renal injury in a model nephropathy: Expression studies in biopsies of type 2 diabetic pa-
of diabetes and hypertension. Kidney Int 62:929–939, 2002 tients. Ren Fail 23:483–493, 2001
40. CHOU E, SUZUMA I, WAY KJ, et al: Decreased cardiac expres- 59. JELKMANN W: Pitfalls in the measurement of circulating vascular
sion of vascular endothelial growth factor and its receptors in endothelial growth factor. Clin Chem 47:617–623, 2001
insulin-resistant and diabetic states: A possible explanation for im- 60. BANKS RE, FORBES MA, KINSEY SE, et al: Release of the angio-
paired collateral formation in cardiac tissue. Circulation 105:373– genic cytokine vascular endothelial growth factor (VEGF) from
379, 2002 platelets: Significance for VEGF measurements and cancer biol-
41. CHIARELLI F, SPAGNOLI A, BASCIANI F, et al: Vascular endothelial ogy. Br J Cancer 77:956–964, 1998
growth factor (VEGF) in children, adolescents and young adults 61. NIELSEN HJ, WERTHER K, MYNSTER T, BRUNNER N: Soluble vascu-
with type 1 diabetes mellitus: Relation to glycaemic control and lar endothelial growth factor in various blood transfusion compo-
microvascular complications. Diabet Med 17:650–656, 2000 nents. Transfusion 39:1078–1083, 1999
42. MCLAREN M, ELHADD TA, GREENE SA, BELCH JJ: Elevated plasma 62. SCHRIJVERS BF, RASCH R, TILTON RG, FLYVBJERG A: High protein-
vascular endothelial cell growth factor and thrombomodulin in induced glomerular hypertrophy is vascular endothelial growth
juvenile diabetic patients. Clin Appl Thromb Hemost 5:21–24, factor-dependent. Kidney Int 61:1600–1604, 2002
1999 63. FLYVBJERG A, SCHRIJVERS BF, DE VRIESE AS, et al: Compensatory
43. DIAMANT M, HANEMAAIJER R, VERHEIJEN JH, et al: Elevated matrix glomerular growth after unilateral nephrectomy is VEGF depen-
metalloproteinase-2 and -9 in urine, but not in serum, are markers dent. Am J Physiol Endocrinol Metab 283:E362–E366, 2002
of type 1 diabetic nephropathy. Diabet Med 18:423–424, 2001 64. PILLEBOUT E, BURTIN M, YUAN HT, et al: Proliferation and remod-
44. MALAMITSI-PUCHNER A, SARANDAKOU A, TZIOTIS J, et al: Serum eling of the peritubular microcirculation after nephron reduction:
levels of basic fibroblast growth factor and vascular endothelial Association with the progression of renal lesions. Am J Pathol
growth factor in children and adolescents with type 1 diabetes 159:547–560, 2001
mellitus. Pediatr Res 44:873–875, 1998 65. KANG DH, JOLY AH, OH SW, et al: Impaired angiogenesis in the
45. HOVIND P, TARNOW L, OESTERGAARD PB, PARVING HH: Elevated remnant kidney model: I. Potential role of vascular endothelial
vascular endothelial growth factor in type 1 diabetic patients with growth factor and thrombospondin-1. J Am Soc Nephrol 12:1434–
diabetic nephropathy. Kidney Int 57 (Suppl 75):S56–S61, 2000 1447, 2001
46. CHATURVEDI N, FULLER JH, POKRAS F, et al: Circulating plasma 66. KANG DH, HUGHES J, MAZZALI M, et al: Impaired angiogenesis in
vascular endothelial growth factor and microvascular complica- the remnant kidney model: II. Vascular endothelial growth factor
tions of type 1 diabetes mellitus: The influence of ACE inhibition. administration reduces renal fibrosis and stabilizes renal function.
Diabet Med 18:288–294, 2001 J Am Soc Nephrol 12:1448–1457, 2001
47. BRAUSEWETTER F, JEHLE PM, JUNG MF, et al: Microvascular per- 67. KANG DH, NAKAGAWA T, FENG L, JOHNSON RJ: Nitric oxide mod-
meability is increased in both types of diabetes and correlates dif- ulates vascular disease in the remnant kidney model. Am J Pathol
ferentially with serum levels of insulin-like growth factor I (IGF-I) 161:239–248, 2002
and vascular endothelial growth factor (VEGF). Horm Metab Res 68. KELLY DJ, HEPPER C, WU LL, et al: Vascular endothelial growth
33:713–720, 2001 factor expression and glomerular endothelial cell loss in the
48. HONKANEN EO, TEPPO AM, GRONHAGEN-RISKA C: Decreased uri- remnant kidney model. Nephrol Dial Transplant 18:1286–1292,
nary excretion of vascular endothelial growth factor in idiopathic 2003
membranous glomerulonephritis. Kidney Int 57:2343–2349, 2000 69. HARPER SJ, DOWNS L, TOMSON CR, et al: Elevated plasma vas-
49. SHULMAN K, ROSEN S, TOGNAZZI K, et al: Expression of vascular cular endothelial growth factor levels in non-diabetic predialysis
permeability factor (VPF/VEGF) is altered in many glomerular uraemia. Nephron 90:341–343, 2002
diseases. J Am Soc Nephrol 7:661–666, 1996 70. STOMPOR T, ZDZIENICKA A, MOTYKA M, et al: Selected growth fac-
50. ZIYADEH FN, HOFFMAN BB, HAN DC, et al: Long-term prevention tors in peritoneal dialysis: Their relationship to markers of inflam-
of renal insufficiency, excess matrix gene expression, and glomeru- mation, dialysis adequacy, residual renal function, and peritoneal
lar mesangial matrix expansion by treatment with monoclonal an- membrane transport. Perit Dial Int 22:670–676, 2002
titransforming growth factor-b antibody in db/db diabetic mice. 71. MATSUMOTO K, OHI H, KANMATSUSE K: Interleukin 10 and inter-
Proc Natl Acad Sci USA 97:8015–8020, 2000 leukin 13 synergize to inhibit vascular permeability factor release
51. TSUCHIDA K, MAKITA Z, YAMAGISHI S, et al: Suppression of trans- by peripheral blood mononuclear cells from patients with lipoid
forming growth factor beta and vascular endothelial growth factor nephrosis. Nephron 77:212–218, 1997
in diabetic nephropathy in rats by a novel advanced glycation end 72. MATSUMOTO K: Interleukin 10 inhibits vascular permeability fac-
product inhibitor, OPB-9195. Diabetologia 42:579–588, 1999 tor release by peripheral blood mononuclear cells in patients with
52. HOSHI S, SHU Y, YOSHIDA F, et al: Podocyte injury promotes pro- lipoid nephrosis. Nephron 75:154–159, 1997
gressive nephropathy in zucker diabetic fatty rats. Lab Invest 73. MATSUMOTO K, OHI H, KANMATSUSE K: Interleukin-4 cooperates
82:25–35, 2002 with interleukin-10 to inhibit vascular permeability factor release
2016 Schrijvers et al: VEGF in renal pathophysiology

by peripheral blood mononuclear cells from patients with minimal- sion in idiopathic membranous nephropathy. Am J Kidney Dis
change nephrotic syndrome. Am J Nephrol 19:21–27, 1999 41:360–365, 2003
74. MATSUMOTO K, OHI H, KANMATSUSE K: Interleukin 12 upregulates 95. NAKAMOTO Y, IMAI H, YASUDA T, et al: A spectrum of clinicopatho-
the release of vascular permeability factor by peripheral blood logical features of nephropathy associated with POEMS syndrome.
mononuclear cells from patients with lipoid nephrosis. Nephron Nephrol Dial Transplant 14:2370–2378, 1999
78:403–409, 1998 96. YAMAMOTO T, KURODA M, NISHIOKA K: Increased serum level of
75. MATSUMOTO K, OHI H, KANMATSUSE K: Effects of interleukin-15 on vascular endothelial growth factor in Crow-Fukase syndrome. Acta
vascular permeability factor release by peripheral blood mononu- Derm Venereol 81:317–318, 2001
clear cells in normal subjects and in patients with minimal-change 97. WATANABE O, MARUYAMA I, ARIMURA K, et al: Overproduction
nephrotic syndrome. Nephron 82:32–38, 1999 of vascular endothelial growth factor/vascular permeability factor
76. MATSUMOTO K, KANMATSUSE K: Interleukin-15 and interleukin-12 is causative in Crow-Fukase (POEMS) syndrome. Muscle Nerve
have an additive effect on the release of vascular permeability 21:1390–1397, 1998
factor by peripheral blood mononuclear cells in normals and in 98. SOUBRIER M, SAURON C, SOUWEINE B, et al: Growth factors and
patients with nephrotic syndrome. Clin Nephrol 52:10–18, 1999 proinflammatory cytokines in the renal involvement of POEMS
77. MATSUMOTO K, KANMATSUSE K: Increased IL-12 release by mono- syndrome. Am J Kidney Dis 34:633–638, 1999
cytes in nephrotic patients. Clin Exp Immunol 117:361–367, 1999 99. KUBO A, NISHITANI Y, MINAMINO N, et al: Adrenomedullin gene
78. MATSUMOTO K, KANMATSUSE K: Transforming growth factor-b1 in- transcription is decreased in peripheral blood mononuclear cells
hibits vascular permeability factor release by T cells in normal of patients with IgA nephropathy. Nephron 85:201–206, 2000
subjects and in patients with minimal-change nephrotic syndrome. 100. AMORE A, CONTI G, CIRINA P, et al: Aberrantly glycosylated IgA
Nephron 87:111–117, 2001 molecules downregulate the synthesis and secretion of vascular
79. MATSUMOTO K, KANMATSUSE K: Augmented interleukin-18 pro- endothelial growth factor in human mesangial cells. Am J Kidney
duction by peripheral blood monocytes in patients with minimal- Dis 36:1242–1252, 2000
change nephrotic syndrome. Am J Nephrol 21:20–27, 2001 101. IRUELA-ARISPE L, GORDON K, HUGO C, et al: Participation of
80. MATSUMOTO K, KANMATSUSE K: Interleukin-18 and interleukin- glomerular endothelial cells in the capillary repair of glomeru-
12 synergize to stimulate the production of vascular permeabil- lonephritis. Am J Pathol 147:1715–1727, 1995
ity factor by T lymphocytes in normal subjects and in patients 102. HA IS, UM EY, JUNG HR, et al: Glucocorticoid diminishes vascu-
with minimal-change nephrotic syndrome. Nephron 85:127–133, lar endothelial growth factor and exacerbates proteinuria in rats
2000 with mesangial proliferative glomerulonephritis. Am J Kidney Dis
81. KONDO S, YOSHIZAWA N, KUSUMI Y, et al: Studies of glomerular 39:1001–1010, 2002
permeability factor (GPF) in focal segmental glomerular sclerosis 103. WADA Y, MORIOKA T, OYANAGI-TANAKA Y, et al: Impairment
and the relationship between GPF and vascular permeability factor of vascular regeneration precedes progressive glomeruloscle-
(VPF). Clin Nephrol 52:278–284, 1999 rosis in anti-Thy 1 glomerulonephritis. Kidney Int 61:432–443,
82. BAKKER WW, BEUKHOF JR, VAN LUIJK WH, VAN DER HEM GK: 2002
Vascular permeability increasing factor (VPF) in IgA nephropa- 104. MASUDA Y, SHIMIZU A, MORI T, et al: Vascular endothelial growth
thy. Clin Nephrol 18:165–167, 1982 factor enhances glomerular capillary repair and accelerates reso-
83. CHEONG HI, LEE JH, HAHN H, et al: Circulating VEGF and TGF- lution of experimentally induced glomerulonephritis. Am J Pathol
beta1 in children with idiopathic nephrotic syndrome. J Nephrol 159:599–608, 2001
14:263–269, 2001 105. YUAN HT, TIPPING PG, LI XZ, et al: Angiopoietin correlates with
84. WEBB NJ, WATSON CJ, ROBERTS IS, et al: Circulating vascular en- glomerular capillary loss in anti-glomerular basement membrane
dothelial growth factor is not increased during relapses of steroid- glomerulonephritis. Kidney Int 61:2078–2089, 2002
sensitive nephrotic syndrome. Kidney Int 55:1063–1071, 1999 106. NANGAKU M, ALPERS CE, PIPPIN J, et al: A new model of renal
85. FAN L, WAKAYAMA T, YOKOYAMA S, et al: Downregulation of vascu- microvascular endothelial injury. Kidney Int 52:182–194, 1997
lar endothelial growth factor and its receptors in the kidney in rats 107. KIM YG, SUGA SI, KANG DH, et al: Vascular endothelial growth
with puromycin aminonucleoside nephrosis. Nephron 90:95–102, factor accelerates renal recovery in experimental thrombotic mi-
2002 croangiopathy. Kidney Int 58:2390–2399, 2000
86. HORITA Y, MIYAZAKI M, KOJI T, et al: Expression of vascular 108. SUGA S, KIM YG, JOLY A, et al: Vascular endothelial growth fac-
endothelial growth factor and its receptors in rats with protein- tor (VEGF121) protects rats from renal infarction in thrombotic
overload nephrosis. Nephrol Dial Transplant 13:2519–2528, microangiopathy. Kidney Int 60:1297–1308, 2001
1998 109. SHAHBAZI M, FRYER AA, PRAVICA V, et al: Vascular endothelial
87. NITTA K, UCHIDA K, HONDA K, et al: Serum vascular endothe- growth factor gene polymorphisms are associated with acute renal
lial growth factor concentration in rapidly progressive glomeru- allograft rejection. J Am Soc Nephrol 13:260–264, 2002
lonephritis. Nephron 80:357–358, 1998 110. PILMORE HL, ERIS JM, PAINTER DM, et al: Vascular endothelial
88. NITTA K, UCHIDA K, KIMATA N, et al: Increased serum levels of growth factor expression in human chronic renal allograft rejec-
vascular endothelial growth factor in human crescentic glomeru- tion. Transplantation 67:929–933, 1999
lonephritis. Clin Nephrol 52:76–82, 1999 111. PILMORE HL, YAN Y, ERIS JM, et al: Time course of upregulation of
89. MATSUMOTO K, KANMATSUSE K: Elevated vascular endothelial fibrogenic growth factors and cellular infiltration in a rodent model
growth factor levels in the urine of patients with minimal-change of chronic renal allograft rejection. Transpl Immunol 10:245–254,
nephrotic syndrome. Clin Nephrol 55:269–274, 2001 2002
90. NOGUCHI K, YOSHIKAWA N, ITO-KARIYA S, et al: Activated mesan- 112. SHIHAB FS, BENNETT WM, YI H, ANDOH TF: Expression of vascular
gial cells produce vascular permeability factor in early-stage endothelial growth factor and its receptors Flt-1 and KDR/Flk-1 in
mesangial proliferative glomerulonephritis. J Am Soc Nephrol chronic cyclosporine nephrotoxicity. Transplantation 72:164–168,
9:1815–1825, 1998 2001
91. PARRY RG, GILLESPIE KM, CLARK AG, MATHIESON PW: Dinu- 113. SHIHAB FS, BENNETT WM, ISAAC J, et al: Angiotensin II regula-
cleotide repeat polymorphisms within the Flt-1 gene in minimal tion of vascular endothelial growth factor and receptors Flt-1 and
change nephropathy. Eur J Immunogenet 26:321–323, 1999 KDR/Flk-1 in cyclosporine nephrotoxicity. Kidney Int 62:422–433,
92. HOLT RC, RALPH SA, WEBB NJ, et al: Steroid-sensitive nephrotic 2002
syndrome and vascular endothelial growth factor gene polymor- 114. SHIHAB FS, BENNETT WM, ISAAC J, et al: Nitric oxide modulates
phisms. Eur J Immunogenet 30:1–3, 2003 vascular endothelial growth factor and receptors in chronic cy-
93. VOGELMANN SU, NELSON WJ, MYERS BD, LEMLEY KV: Urinary closporine nephrotoxicity. Kidney Int 63:522–533, 2003
excretion of viable podocytes in health and renal disease. Am J 115. ALVAREZ ARROYO MV, SUZUKI Y, YAGUE S, et al: Role of endoge-
Phsyiol Renal Physiol 285:F40–F48, 2003 nous vascular endothelial growth factor in tubular cell protection
94. SIVRIDIS E, GIATROMANOLAKI A, TOULOUPIDIS S, et al: Platelet en- against acute cyclosporine toxicity. Transplantation 74:1618–1624,
dothelial cell adhesion molecule-1 and angiogenic factor expres- 2002
Schrijvers et al: VEGF in renal pathophysiology 2017

116. KANG DH, KIM YG, ANDOH TF, et al: Post-cyclosporine-mediated lupus erythematosus and primary antiphospholipid syndrome.
hypertension and nephropathy: Amelioration by vascular Lupus 11:21–24, 2002
endothelial growth factor. Am J Physiol Renal Physiol 280:F727– 124. LI CG, REYNOLDS I, PONTING JM, et al: Serum levels of vascular
F736, 2001 endothelial growth factor (VEGF) are markedly elevated in pa-
117. YOSHIDA T, KUWAHARA M, MAITA K, HARADA T: Immunohisto- tients with Wegener’s granulomatosis. Br J Rheumatol 37:1303–
chemical study on hypoxia in spontaneous polycystic liver and 1306, 1998
kidney disease in rats. Exp Toxicol Pathol 53:123–128, 2001 125. DVORAK HF: Vascular permeability factor/vascular endothelial
118. BELLO-REUSS E, HOLUBEC K, RAJARAMAN S: Angiogenesis in growth factor: A critical cytokine in tumor angiogenesis and a po-
autosomal-dominant polycystic kidney disease. Kidney Int 60:37– tential target for diagnosis and therapy. J Clin Oncol 20:4368–4380,
45, 2001 2001
119. KANELLIS J, MUDGE SJ, FRASER S, et al: Redistribution of cyto- 126. VAN NIEUW AMERONGEN GP, VAN HINSBERGH VWM: Targets
plasmic VEGF to the basolateral aspect of renal tubular cells in for pharmacological intervention of endothelial hyperperme-
ischemia-reperfusion injury. Kidney Int 57:2445–2456, 2000 ability and barrier function. Vascul Pharmacol 39:257–272,
120. KANELLIS J, PAIZIS K, COX AJ, et al: Renal ischemia-reperfusion 2002
increases endothelial VEGFR-2 without increasing VEGF or 127. FERRARA N: Role of vascular endothelial growth factor in physio-
VEGFR-1 expression. Kidney Int 61:1696–1706, 2002 logic and pathologic angiogenesis: Therapeutic implications. Semin
121. HALTIA A, SOLIN ML, JALANKO H, et al: Mechanisms of proteinuria: Oncol 29:10–14, 2002
Vascular permeability factor in congenital nephrotic syndrome of 128. THE EYETECH STUDY GROUP: Preclinical and phase 1A clinical eval-
the Finnish type. Pediatr Res 40:652–657, 1996 uation of an anti-VEGF pegylated aptamer (EYE001) for the
122. NISHITANI Y, KUBO A, IWANO M, et al: Imbalance between treatment of exudative age-related macular degeneration. Retina
interleukin-6 and adrenomedullin mRNA levels in peripheral 22:143–152, 2002
blood mononuclear cells of patients with lupus nephritis. Clin Exp 129. PUPILLI C, LASAGNI L, ROMAGNANI P, et al: Angiotensin II stim-
Immunol 124:330–336, 2001 ulates the synthesis and secretion of vascular permeability fac-
123. NAVARRO C, CANDIA-ZUNIGA L, SILVEIRA LH, et al: Vascular en- tor/vascular endothelial growth factor in human mesangial cells. J
dothelial growth factor plasma levels in patients with systemic Am Soc Nephrol 10:245–255, 1999

Вам также может понравиться