Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 19.
Published in final edited form as: Cell Death Differ. 2008 Jan 25;15(4):650–659. doi: 10.1038/sj.cdd.4402313

The VHL tumor suppressor and HIF: insights from genetic studies in mice

PP Kapitsinou 1, VH Haase 1,*
PMCID: PMC3799983  NIHMSID: NIHMS516580  PMID: 18219317

Abstract

The von Hippel–Lindau tumor suppressor gene product, pVHL, functions as the substrate recognition component of an E3-ubiquitin ligase, which targets the oxygen-sensitive α-subunit of hypoxia-inducible factor (HIF) for rapid proteasomal degradation under normoxic conditions and as such plays a central role in molecular oxygen sensing. Mutations in pVHL can be found in familial and sporadic clear cell carcinomas of the kidney, hemangioblastomas of the retina and central nervous system, and pheochromocytomas, underscoring its gatekeeper function in the pathogenesis of these tumors. Tissue-specific gene targeting of VHL in mice has demonstrated that efficient execution of pVHL-mediated HIF proteolysis under normoxia is fundamentally important for survival, proliferation, differentiation and normal physiology of many cell types, and has provided novel insights into the biological function of individual HIF transcription factors. In this review, we discuss the role of HIF in the development of the VHL phenotype.

Keywords: von Hippel–indau (VHL) tumor suppressor, hypoxia-inducible factor (HIF), conditional knockout mice, renal cell cancer, hemangiomas, vascular tumors


Germ-line mutations in the von Hippel–Lindau tumor (VHL) suppressor can be found in patients with VHL disease, a rare familial tumor syndrome characterized by the development of highly vascularized tumors in multiple organs. Major clinical manifestations of VHL disease include hemangioblastomas of the retina and central nervous system (CNS), renal cysts and renal cell carcinoma of the clear cell type (CC-RCC), pancreatic cysts and tumors, as well as pheochromocytomas.1 The VHL tumor suppressor is also mutated in the majority of sporadic CC-RCCs,2 highlighting its critical and central role in the regulation of cell growth and differentiation of epithelial kidney cells. Although the molecular function of the VHL gene product, pVHL, was initially not known when it was first identified by positional cloning in 1993,3 observations that oxygen-dependent regulation of hypoxia-inducible genes was lost in VHL-deficient cell lines suggested a role for pVHL in oxygen sensing.46 In a seminal paper, Maxwell et al.7 showed that pVHL was critical for targeting the α-subunit of hypoxia-inducible factor (HIF) for oxygen-dependent proteolysis, thus providing a direct molecular link between VHL-associated tumorigenesis and oxygen sensing via HIF. HIFs belong to the PAS (Per-arylhydrocarbon receptor nuclear translocator (ARNT)-Sim) family of basic helix-loop-helix (bHLH) transcription factors and bind DNA as heterodimers. They consist of an oxygen-sensitive α-subunit and a constitutively expressed β-subunit, also known as ARNT or HIF-β. pVHL associates with the elongins B and C, cullin2 and Rbx812 and functions as the substrate recognition component of an E3-ubiquitin ligase that ubiquitylates HIF-α.1319 All three HIF α-subunits, HIF-1α, HIF-2α and HIF-3α interact with pVHL.7,20 This pVHL–HIF-a interaction is highly conserved between species, requires iron- and oxygen-dependent hydroxylation of specific proline residues (Pro402 and Pro564 in human HIF-1α; Pro405 and Pro531 in human HIF-2α) within the oxygen-dependent degradation domain of HIF-α and is necessary for the execution of HIF proteolysis under normoxia.2127 Therefore, absence of pVHL results in HIF-α stabilization, increased HIF transcriptional activity and upregulation of HIF target genes, such as vascular endothelial growth factor (VEGF), glucose transporter 1 and erythropoietin (EPO) irrespective of oxygen levels.

To understand the role of pVHL-mediated HIF proteolysis in normal tissue physiology, tumorigenesis and during development, we have used cell-type-specific gene targeting based on Cre recombinase (Cre)/loxP-mediated recombination. In this review, we will summarize the findings from a variety of developmental and physiological studies in conditional VHL and HIF knockout mice and provide insights into the biological function of individual HIF transcription factors in a VHL-deficient background.

Normoxic Stabilization of HIF-α in VHL-Deficient Tissues

pVHL is a master regulator of HIF-α proteolysis, and genetic inactivation of the VHL gene results in HIF-α stabilization and increased activity of HIF transcription factors. The most extensively studied PAS family transcription factors that are targeted by pVHL are HIF-1 and HIF-2 (here collectively referred to as HIF). HIFs regulate gene expression through interaction with hypoxia response elements, specific DNA recognition sequences that are located in hypoxia enhancer regulatory regions and bind HIF heterodimers.28 HIF-1 and HIF-2 have distinct cell type- and tissue-specific biological functions and only overlap partially with regard to their target genes. For example, genes encoding glycolytic enzymes appear to be predominantly controlled by HIF-1,29 whereas HIF-2 appears to be the main regulator of VEGF and EPO in tissues that express both HIF-1 and HIF-2.3033 Target gene preference may be the result of tissue-specific interactions with other nuclear factors, differential interactions with transcriptional cofactors or a reflection of tissue- and cell-type-dependent differences in the ratios of HIF-α protein levels (for a review on this topic see Wenger et al.28).

In addition to heterodimerization with HIF-β, which results in the formation of a bHLH transcription factor that mediates the canonical hypoxia response, stabilization of HIF-α subunits has also been shown to regulate biological processes through functional interaction with other non-PAS domain proteins. These include, among others, tumor suppressor protein p53 and the c-Myc proto-oncogene.3436 A more recent example is the ability of HIF-1α to associate with the intracellular domain of Notch (Notch ICD), thereby increasing the expression of Notch-target genes, such as Hey and Hes.37 This and other HIF-α/non-PAS domain protein interactions have to be considered when interpreting experimental phenotypes that result from VHL gene deletion in the laboratory mouse.

A second hypoxic switch operates in the carboxy-terminal transactivation domain of HIF-α with the hydroxylation of an asparagine residue.38 During hypoxia, asparagine hydroxylation is blocked and CBP/p300 recruitment is facilitated enabling increased levels of transcription. Factor-inhibiting HIF (FIH) hydroxylates the asparagine residue at position Asn803 in human HIF-1α, which corresponds to asparagine Asn851 in HIF-2α.39,40 Inhibition of FIH can result in increased HIF target gene expression even under severe hypoxia or in certain VHL-deficient cell lines.41

Biological Functions not Involving HIF

Aside from mediating HIF-α proteolysis, pVHL is involved in extracellular matrix (ECM) assembly matrix turnover,4247 the regulation of intracellular junctions,48 NF-κB signaling,49 the regulation of c-Met receptor responsiveness to hepatocyte growth factor (HGF) involving β-catenin45,50 and the regulation of p53 transcriptional activity by suppressing Mdm2-mediated ubiquitination and nuclear export.51 Furthermore, pVHL has been shown to regulate microtubule stability and cilia maintenance5255 and controls the activity of plant homeodomain protein Jade-1,56,57 and atypical protein kinase C isoforms.5862 Other pVHL targets include a KRAB-A domain protein, VHLak, repressing HIF transcriptional activity,63 de-ubiquitinating enzymes,64 the large subunit of RNA polymerase II65 and the RNA-binding protein hnRNP A2.66 How these HIF-independent pVHL functions and recently discovered protein interactions exactly contribute to the initiation and progression of VHL-associated tumorigenesis is unclear and requires further investigation.

pVHL during Embryonic Development

Genetic inactivation of pVHL in the mouse germ line results in death of the embryo during mid-gestation. Although placenta and embryo appear normal until embryonic day (E) E9.5, placentae from VHL-deficient embryos lack a properly developed syncytiotrophoblast and labyrinth and show evidence of hemorrhage by E11.5–12.5, suggesting that abnormal placental vascularization has led to the embryo’s demise.67 This phenotype appears to be largely mediated by HIF, since germ-line inactivation of HIF prolyl-hydroxylase domain (PHD) protein 2 resulted in similar, however, not identical pathology,68 indicating HIF-independent functions of either pVHL and/or PHD2 during placental development. pVHL is not only essential for normal placental development but also plays a critical role in the development, growth and differentiation of many other tissues. For example, tissue-specific inactivation of pVHL in neuro-epithelial progenitor cells resulted in abnormal neuronal differentiation and embryonic lethality during late gestation (VH Haase, unpublished data), and chondrocyte-specific inactivation of pVHL in the growth plate caused stunted bone growth, most likely a consequence of a HIF-dependent increase in cell cycle inhibitor p57kip2.69 In contrast, HIF-1α inactivation in chondrocytes resulted in a lack of p57kip2-mediated growth arrest, followed by massive apoptosis.70

VHL Disease Manifestations in Mice with Germ-line Mutations

Patients with VHL disease develop highly vascularized tumors in multiple organ systems, hemangioblastomas of the retina and CNS, CC-RCC and pheochromocytomas being the most prominent clinical manifestations.1 VHL disease is grouped into two subtypes depending on the presence or absence of pheochromocytoma. Different clinical subtypes are associated with specific VHL mutations, which result in distinct functional and biochemical properties of the mutated VHL gene product (for an overview see Neumann and Bender71, Clifford et al.72 and Hoffman et al.73). Clinical manifestations of VHL disease occur relatively early in life, usually between 10 and 40 years of age, whereas sporadic tumors with VHL mutations, such as CC-RCC present later in life. In addition to tumor formation, mutations in the VHL gene can result in the development of polycythemia.74 Congenital Chuvash polycythemia is a rare endemic disease in central European Russia and is associated with a specific mutation of VHL codon 200 (C598T ⇒ Arg200Trp). This mutation is transmitted in an autosomal-recessive manner, and affected individuals, who are homozygous for the mutation, are not predisposed to the development of typical VHL-associated tumors.75

In mice, germ-line loss of one VHL allele (VHL +/−) results in the development of cavernous liver hemangiomas,76 which is a rare manifestation of VHL disease in human.77,78 This phenotype is strongly dependent on the genetic background of heterozygous mice, as the incidence of liver hemangiomas in BALB/c mice was 88%, but only 18% in a C57BL/6 background, most likely reflecting polymorphic differences in modifier genes.79

Although the molecular mechanism for this liver phenotype has not been systematically investigated, it is most likely that, following Knudson’s two-hit hypothesis, inactivation of the remaining VHL wild-type allele in hepatocytes resulted in the formation of cavernous hemangiomas through an increase in HIF-dependent vascular growth factor production. Hepatocyte-specific inactivation of VHL using the Albumin-Cre and phosphoenolpyruvate carboxykinase (PEPCK)-Cre trans-genes (Figure 1 and Table 1) phenocopies the liver pathology found in heterozygotes and suggests that liver hemangiomas in VHL heterozygotes are the result of pVHL loss in hepatocytes and not in other liver cell types, such as endothelial cells. Endothelial cells, which express wild-type VHL in this model, respond to uncontrolled and constitutive production of hepatocyte-derived vascular growth factors with proliferation. This is mechanistically similar to human VHL-associated hemangioblastomas, in which ‘stromal cells’ and not endothelial cells are VHL-deficient and represent the neoplastic component of these tumors.94 The histogenetic origin of stromal cells is uncertain; however, correlative evidence suggests that they may be derived from angiogenic progenitor cells.95,96

Figure 1.

Figure 1

Hepatic vascular tumors in mice that lack pVHL in hepatocytes. (a) Gross photograph of a liver with multiple cavernous hemangiomas (arrows). (b) Histological features of cavernous liver hemangiomas. A paraffin-embedded tissue section stained with H&E is shown. Asterisks depict endothelial cell-lined, blood-filled cavities and areas of hemorrhage. VHL-associated liver hemangiomas are typically associated with macro- and microvesicular hepatocellular accumulation of neutral lipids,76 here depicted by arrows. Lipid accumulation is also a feature of stromal cells, the neoplastic component of VHL-associated central nervous system hemangioblastomas95,96

Table 1.

Phenotypes generated by cell-type-specific inactivation of pVHL in mice

References Cre transgenic
(promoter)
Cell type Phenotype
Kidney
Ding et al.80 NPHS2 (podocin) Podocytes Acute nephritis (4 weeks) with hematuria, proteinuria and renal insufficiency;
features characteristic of pauci-immune RPGN (crescentic glomerulonephritis with prominent segmental fibrin deposition and fibrinoid necrosis, no immune deposits); absence of circulating ANCA antibodies; both HIF-1α and HIF-2α
protein levels were increased; upregulation of HIF-dependent genes (including CXCR4) as well as RPGN-associated genes; de novo expression of CXCR4
Brukamp et al.81 NPHS2 (podocin) Podocytes No prominent disease pattern in vivo; preserved glomerular development but
glomerulomegaly at a young age and occasional glomerulosclerosis; no
significant proteinuria; increased podocyte apoptosis in vitro
Steenhard et al.82 NPHS2 (podocin) Podocytes Initially normal glomerular development; at 16 weeks ultrastructural changes,
such as foot process broadening, widespread irregular GBM thickening with
subepithelial hump formation and focal subendothelial delamination; proteinuria
Rankin et al.83 PEPCK
(phosphoenolpyruvate carboxykinase)
Proximal renal
tubule
Renal cysts at low frequency; inactivation of ARNT, but not HIF-1 α suppressed
the development of renal cysts
Liver
Haase et al.76 Albumin Hepatocytes Growth deficiency, angiectasis, hemangiomas, endothelial cell proliferation,
severe hepatic steatosis (neutral fat accumulation in hepatocytes),
inflammatory cell infiltration, erythrocytosis from increased liver EPO
production; HIF-2 is the main mediator of this phenotype; HIF-1 regulates
glycolytic gene expression
Rankin et al.31
Rankin et al.32
Peyssonnaux
et al.84
Albumin
Hepatocytes Relative iron deficiency with hypochromia, poikilocytosis, microcytosis,
decreased total iron and ferritin levels; spleen also strongly iron deficient;
decreased hepcidin mRNA and protein levels (ARNT dependent); hepcidin
downregulation specifically due to the stabilization of HIF; significantly elevated
IL-6 and IL-1 levels; IL6- or IL1-mediated stimulation of hepcidin expression is
subordinate to the suppression by HIF; increased ferroportin expression in
brush border enterocytes of the duodenum, Kupffer cells and hepatocytes
Rankin et al.31,32 PEPCK
(phosphoenolpyruvate carboxykinase)
Hepatocytes Liver hemangiomas, polycythemia; HIF-2α is required for the development of
polycythemia and inactivation of HIF-2α is sufficient to suppress hepatic EPO
expression in VHL-KO livers despite increased HIF-1 activity
Immune system
Neumann et al.85 Lck (lymphocyte protein tyrosine kinase) Thymocytes Diminished maximal Cai2+ together with retarded initial rates of Cai2+ elevation in
response to CD3/CD4 co-ligation;
Ca2+signaling was completely restored in VHL−/−/HIF-1α−/− double KO
thymocytes; stabilization of HIF-1α increases flux of Ca2+from the cytoplasm to
the mitochondria and results in increased protein levels of SERCA2 (Ca2+pump that refills ER stores); overall SERCA-mediated Ca2+transport is increased;
both hypoxic and non-hypoxic-mediated HIF-1 α stabilization may minimize the
strength and duration of Ca2+signaling by means of acceleration of cytoplasmic
Ca2+clearance
Biju et al.86 Lck (lymphocyte protein tyrosine kinase) Thymocytes Small, highly vascularized thymi; increased apoptosis in CD4/CD8 double-
positive cells involving a caspase8-dependent mechanism; phenotype is HIF-1
dependent.
Cramer et al.87 LysM (lysozyme M) Myeloid cells Increase in inflammatory response through HIF-1
Epithelium
Karhausen et al.88 Fabp (fatty acid-binding protein) Colonic
epithelial
cells
Increased expression of HIF-regulated barrier protective factors such as
intestinal trefoil factor; protection from inflammatory bowel disease clinically;
accumulation of glycogen in luminal epithelial cells
Seagroves et al.89a WAP (whey acidic protein) Mammary
gland
epithelium
Abnormal differentiation of gland epithelium during lactation in multiply bred
mutants, collapsed alveoli, little or no milk production, enlarged blood vessels,
no hyperplasia or tumors
Boutin et al.90a K14 (keratin 14) Epidermis Increased dermal vasculature, higher metabolic rate, low weight, early death,
erythrocytosis
Vascular system
Tang et al.91 Tie2 Endothelial
cells
Intrauterine death with hemorrhages at E12.5; loss of HIF-1α did not rescue the
embryonic lethality; endocardial collapse, abnormal yolk sac vascular
organization, reduced fibronectin deposition around vitelline vessels, defective
extracellular fibronectin matrix assembly, defective vasculogenesis in the
placental labyrinth, dilated vessels and reduced vessel complexity; increased
permeability of the endothelial monolayer; impaired migration and impaired
adhesion partially rescued with external fibronectin; regulation of fibronectin
assembly independent of HIF-1
Connective tissue
Wang et al.92 OC (osteocalcin) Osteoblasts Bone modeling is increased within the first week of life but appears to decline at
latter stages of development; extremely dense, heavily vascularized long
bones; progressively increased vessel numbers with age; trabecular separation
reduced; no detectable changes in calvarial bone morphology; upregulation of
HIF-1 and HIF-2 and VEGF, serum level of VEGF was not elevated; conversely,
inactivation of HIF-1α in the osteoblast produced the reverse phenotype, that is,
thinner and less vascularized bones
Pfander et al.69 ColII (collagen II) Growth plate/
chondrocytes
Severe dwarfism, severe decrease in chondrocyte proliferation, upregulation of
HIF-1-dependent p57kip2, increased extracellular matrix deposition
Mosaic and neurons
Hong et al.93 ROSA26-tamoxifen-
inducible ER-Cre
Mosaic Distinct changes at E14.5; embryonic lethality; small, pale embryos; extensive
hemorrhage; vascular defects and liver damage in the embryos, but no major
placental defects prior to death; no significant increase in apoptosis or decrease
in proliferation
Ma et al.79 β-actin-tamoxifen-
inducible ER-Cre
Mosaic Hepatic vascular tumors; angiectasis in heart, liver, pancreas, lung, kidney;
infertility, abnormal spermatogenesis
Haase et al.,
unpublished
observation
Nestin Neuro-epithelial
progenitor cells
Abnormal neuronal differentiation, embryonic lethality

A compilation of mouse phenotypes organized by organ system and cell types, which were made pVHL deficient, is presented. The gene regulatory elements used for the construction of Cre transgenes to induce cell-type-specific inactivation of VHL are listed in the second column.

a

Meeting abstract; ANCA, anti-neutrophilic cytoplasmic antibody; CXCR4, chemokine receptor 4; RPGN, rapidly progressive glomerulonephritis

Another major visceral manifestation of VHL disease is the development of kidney cysts, which can occur in up to 60% of patients with VHL germ-line mutations.1,97 Renal cysts, which occur more frequently than CC-RCC, are considered preneoplastic precursor lesions of CC-RCC. In VHL + /− mice renal cysts were only found at extremely low frequency (<5%) by Haase et al.76 and Kleymenova et al.98 In both reports, CC-RCCs were not observed, nor were CNS hemangioblastomas or retinal angiomas. Treatment with streptozotocin did not result in increased susceptibility to renal carcinogenesis, but increased the incidence of hemangiomas and hemangiosarcomas in the liver and other organs such as the uterus and ovaries.98

Although erythrocytosis is seen in a small fraction of patients with VHL disease,99,100 elevated hematocrits are not found in VHL + /− mice. Mice homozygous for a VHL mutation at codon 200, which does not result in embryonic lethality when bred to homozygosity, developed mild HIF-2-dependent erythrocytosis, thus mimicking features of human Chuvash polycythemia.101

pVHL and Renal Cell Cancer

A molecular hallmark of the most common form of kidney cancer, CC-RCC, is mutated pVHL. In contrast to patients with VHL disease, who transmit germ-line mutations, patients with sporadic CC-RCC have acquired somatic mutations in both copies of the VHL gene. VHL-associated CC-RCCs are often preceded by multifocal and bilateral renal cysts, which can be found in up to 60% of patients with VHL disease and can sometimes mimic polycystic kidney disease.1,97 VHL-associated renal cysts can be malignant or benign, and should be viewed as preneoplastic lesions. Renal cystogenesis in general is associated with altered signaling through the primary cilium, a microtubule-based organelle, which is found in many cell types such as neurons, photoreceptors, fibroblasts and others, and predominantly functions as a luminal flow sensor on renal epithelial cells and regulates renal tubular cell proliferation.102 More recent studies have proposed that pVHL is essential for cilium maintenance.5255,103 pVHL was reported to cooperate with GSK3β in an interlinked signaling pathway that maintains the primary cilium.53 Schermer et al.54 demonstrated that pVHL localizes to the monocilia of kidney cells, interacts with Par3-Par6-aPKC and controls ciliogenesis via coordinated extension of microtubules toward the cell periphery and Esteban et al.103 proposed a role for HIF in cilium maintenance. The latter study showed that pVHL inactivation is associated with the loss of the primary cilium in VHL-defective cell lines and in kidney cysts found in patients with VHL disease, and that reconstitution of cell lines with wild-type pVHL restored primary cilia in a HIF-dependent manner. Although CC-RCCs are traditionally believed to arise from the proximal renal tubule,97,104 their histogenetic origin remains controversial. Recent immunohistochemical studies suggested that CC-RCC originates from distal nephron segments, which express Tamm–Horsfall protein (THP).105 While certain features of VHL-deficient tumors, for example, their highly vascular nature resulting from increased VEGF production, can be directly linked to constitutive HIF stabilization, renal carcinogenesis is more difficult to understand and cannot be explained by activated HIF signaling alone. However, loss of pVHL function and subsequent HIF-α stabilization represent the earliest detectable molecular events in renal tumorigenesis.105 Esteban et al.106 showed that pVHL inactivation is associated with downregulation of intercellular adhesion molecule E-cadherin in pre-cancerous lesions and that both HIF-1 and HIF-2 contributed to decreased E-cadherin expression with HIF-2 mediating a more potent suppression in CC-RCC cell lines. Similarly, Evans et al.107 demonstrated that knockdown of pVHL resulted in E-cadherin suppression via HIF-dependent induction of E2 box-dependent transcriptional repressors Snail and SIP1, and Krishnamachary et al.108 reported that HIF-1 activation in VHL-deficient cells downregulated E-cadherin, led to the loss of cell–cell adhesion and promoted epithelial to mesenchymal transition through the induction of transcriptional repressors TCF3, ZFHX1A and ZFHX1B/SIP1. Therefore, cellular changes, such as loss of intercellular junctions and epithelial de-differentiation involving HIF-dependent as well as HIF-independent molecular path-ways48,106108 in addition to HIF-dependent and -independent alterations in p53 or NF-κB activity,34,35,49,51 HGF signaling,45,50,109 and modifications in ECM turnover and re-modeling4247 create the molecular environment for the development CC-RCC, which most likely requires additional genetic events. The importance of HIF activation in CC-RCC pathogenesis and growth is furthermore underscored by experimental and clinical studies, which demonstrated that inhibition of HIF-α translation by pharmacological targeting of mTOR correlated with reduced tumor growth,110 and that increased expression of certain HIF target genes, such as CXCR4, as well as E-cadherin suppression was associated with disease progression.111,112 With regard to the contribution of individual HIF-α homologs to renal tumor development, it is of interest that a substantial number of VHL-defective CC-RCC cell lines do not express HIF-1α, but express HIF-2α.7 Furthermore a bias toward HIF-2α expression was also found in clinical CC-RCC samples with confirmed VHL defect.113 This is in contrast to normal, non-transformed renal epithelial cells, in which HIF-2α is usually not detectable during hypoxia/ ischemia.114 Thus, VHL-associated tumor development may depend on a shift in the ratio of HIF-1α versus HIF-2α levels toward an increase in HIF-2α. In support of this hypothesis are studies in CC-RCC cell lines, which suggest that HIF-2 in contrast to HIF-1 is oncogenic and is able to override pVHL’s tumor suppressor function.115118 HIF-2 has been proposed to preferentially regulate molecular pathways critical for renal cell growth, such as signaling through the TGF-α/epidermal growth factor receptor pathway, cyclin D1 and the c-Myc proto-oncogene.119124 Taken together, there is substantial evidence that HIF-1 and HIF-2 have diverse functions with regard to VHL renal tumorigenesis, which could be exploited therapeutically, and may be strongly context dependent.125 Not much is known about the role of HIF-3α in VHL-associated tumorigenesis. Recent studies, however, have suggested that a specific splice form of HIF-3α, HIF-3α4, inhibits renal tumor growth through abrogation of HIF-2 signaling.126

To overcome embryonic lethality and to generate a mouse model of VHL-associated CC-RCC, we have used Cre/loxP-mediated gene targeting to inactivate pVHL in adult renal tissues. In the conditional VHL allele, promoter and exon 1 are flanked by loxP sites and are deleted upon Cre-mediated recombination, resulting in a mutated VHL allele that is transcriptionally inactive.76 Similar to our approach, Ma et al.79 generated a conditional VHL allele in which exons 2 and 3 were floxed. To specifically target renal epithelial cell types from which CC-RCCs are thought to originate, we generated transgenic mice, which express Cre in proximal and distal renal tubule segments under control of either a mutated version of the rat PEPCK or the THP promoter83 (also EB Rankin et al., unpublished observations). PEPCK-Cre mutant mice developed renal cysts at a frequency of ∼20%, but CC-RCC was not observed (Figure 2). Renal cyst development in PEPCK-Cre mutant was dependent on intact HIF signaling, but not on HIF-1α, as cysts were not observed in VHL/ARNT double knockout mice and HIF-1α was not required for cyst formation.83 Activation of HIF signaling has also been suggested to promote renal cystogenesis in a mouse model of fumarate hydratase deficiency, which leads to HIF prolyl-hydroxylase inhibition and HIF-α stabilization,127 indicating that HIF-mediated renal cystogenesis is not dependent on VHL status. In contrast to pVHL inactivation with PEPCK-Cre transgenics, inactivation of pVHL using THP-Cre, which targets the medullary thick ascending loop of Henle (mTAL) and the early distal tubule, did not produce renal cysts. CC-RCCs were not observed in THP-Cre mutant animals. Renal tumors were also not found by Ma et al.79 when an inducible β-actin promoter-driven Cre was used to generate mice which lacked pVHL in a mosaic pattern. The absence of renal tumorigenesis in VHL-deficient mice in conjunction with genetic data from humans suggests that transformation of renal cysts into CC-RCC most likely requires additional genetic events such as mutations in other tumor suppressor genes or oncogenes (Figure 3).

Figure 2.

Figure 2

Mice that lack pVHL in the proximal renal tubule develop tubular and glomerular cysts. (a) Gross in situ photograph of a kidney from a male PEPCK-Cre mutant with two macroscopically visible cysts (arrows). The PEPCK-Cre transgene was used to inactivate pVHL in the proximal renal tubule. Genetic analysis with compound knockout mice demonstrated that this phenotype was HIF dependent.83 (b-d) Histological features of cystic changes found in H&E-stained paraffin sections from VHL-deficient kidneys. A large cortical cyst (b), a cluster of renal tubular cysts lined by cuboidal epithelium (asterisks in c) and an area of glomerular cysts (number sign in d) are shown. CC-RCC or dysplastic changes in cysts were not found in VHL-deficient kidneys. Magnification is × 100

Figure 3.

Figure 3

Schematic illustrating the role of pVHL and HIF in the development of the VHL phenotype. While activation of HIF signaling is required and sufficient for vascular tumor development, renal cystogenesis in humans may be a result of HIF activation ± loss of HIF-independent functions of pVHL. CC-RCC development most likely requires additional mutations in other tumor suppressor genes or in certain oncogenes. (a) Gross photography of a liver hemangioma found in a VHL mutant mouse. (b, c) Human renal cell carcinoma of the clear cell type (CC-RCC); (b) gross photography; (c) PAS stain of CC-RCC, clear cells are depicted by arrows, magnification × 400; images (b, c) were kindly provided by Dr. John Tomaszewski, Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA

VHL Phenotypes in Non-Renal Tissues: is it All HIF?

To study the role of pVHL in growth, differentiation and normal tissue physiology, we have used different Cre transgenic lines to inactivate pVHL in multiple tissues. Table 1 summarizes clinical phenotypes that we and others have observed. Inactivation of pVHL results in abnormal cellular differentiation and growth causing significant organ pathology in many tissues, one common feature being increased vascularity. Although HIF-mediated stimulation of vascular growth factor production can explain increased vascularity, the effects of pVHL gene deletion on cellular growth and differentiation, metabolism and organ function are not as easily understood. In a non-RCC tumor model, Mack et al.128 found significant growth retardation in teratocarcinomas derived from VHL−/−embryonic stem cells. These findings were surprising, but are in concordance with observations made by Carmeliet et al.129 who showed that HIF-1 α-deficient teratocarcinomas had a growth advantage compared to wild type. However, growth suppression in VHL−/− teratocarcinomas may not be entirely dependent on HIF-1, as introduction of a mutated pVHL species (Y112H) into the VHL−/− background remained growth suppressive despite restoration of HIF-α regulation,130 suggesting HIF-independent functions of pVHL in this model.

To determine the role of individual HIF transcription factors in the development of the VHL phenotype in mice, we generated tissue-specific pVHL knockout mice, which lack HIF-1 α and/or HIF-2α or HIF-1 β(ARNT). Using this compound knockout approach, Biju et al.86 was able to show that a caspase-8-dependent pro-apoptotic phenotype in VHL−/− thymocytes was completely HIF-1 mediated. HIF-2α, on the other hand, although expressed in thymocytes, was found to be non-functional and inactivation of HIF-1 α alone was sufficient to completely suppress VEGF mRNA levels and the vascular phenotype in VHL−/− thymi.86 This is contrast to the VHL-deficient liver, where inactivation of HIF-1 α, expressed and functional in hepatocytes, did not affect VHL-associated vascular tumorigenesis, while inactivation of HIF-2α was sufficient to prevent the development of cavernous hemangiomas (EB Rankin and VH Haase, submitted). This finding is consistent with the observation that angiogenic gene expression in hepatocytes, similar to the regulation of EPO, is strongly HIF-2 dependent32 (also EB Rankin and VH Haase, submitted) and illustrates, that although HIF-1 and HIF-2 have the capability to regulate the same target genes, preferential transcriptional activation by HIF-1 or HIF-2 in the setting of VHL deficiency and/or hypoxia is tissue- or context-dependent.120 Whether this requires tissue-specific transcriptional co-activators or repressors, certain DNA or protein modifications or other signaling events warrants further investigation.131 Whatever the underlying mechanisms, clinical studies have also demonstrated a correlation between HIF-2α expression and the development of VHL-associated angiogenic lesions,132 suggesting that pharmacological targeting of HIF-2 may by an effective therapy for the treatment of these tumors.

pVHL- and pVHL/ARNT-deficient liver tissues are histologically similar and direct comparison of gene expression changes has not revealed major differences31 (also VH Haase et al., unpublished observation), suggesting that the effects of pVHL gene deletion in the liver are largely HIF mediated. Indeed, forced expression of non-degradable HIF-1α and HIF-2α phenocopied the VHL phenotype in the liver and skin133 (Table 2a). Similarly, genetic inactivation of PHD2 in the mouse germ line resulted in placental changes that resembled, but were not identical to those observed in VHL knockout mice.68 PHD2, one of three major mammalian HIF prolyl-hydroxylases, is essential for HIF-α degradation under normoxia,68,134,135 while PHD3 seems to be important for hydroxylation of HIF-α during re-oxygenation.136 Analysis of tissue-specific PHD knockout mice and direct comparison with VHL phenotypes is therefore likely to provide novel insights into HIF-dependent and -independent functions of pVHL (see Table 2b).

Table 2.

Mouse phenotypes resulting from HIF activation in tissues with wild-type VHL

References Cre transgenic
(promoter)
Cell type Phenotype
(a) Liver
Kim et al.133 Albumin; HIF1dPA
(non-degradable HIF-1α)
Hepatocytes Normal lifespan, normal appearing liver but with fine vacuolization
of hepatocytes; moderate lipid accumulation (microvesicular pattern); normal proliferation
Albumin; HIF2dPA
(non-degradable HIF-2α)
Hepatocytes Death at 6–8 weeks, marked hepatomegaly, polycythemia from
increased liver EPO production; angiectasis; minimal
vacuolization of hepatocytes; increased proliferation
Albumin; HIF1dPA/
HIF2dA
Hepatocytes Death at 6–8 weeks; marked hepatomegaly; angiectasis; micro
and macrovesicular hepatic steatosis (histologically identical to
Albumin-Cre VHL−/− mutants), increased proliferation;
polycythemia
Skin
Kim et al.133 K14 (keratin 14);
HIF1dPA
Basal keratinocytes No gross skin phenotype
K14 (keratin 14);
HIF2dPA
Basal keratinocytes Formation of non-leaky blood vessels; partial alopecia; runting;
epidermal proliferation; weight loss
(b) Early embryo and mosaic
Takeda et al.68 EIIa (adenovirus promoter) General deleter,
early embryo prior to
implantation
PHD2−/−:
Embryonic lethality occurred between E12.5–E14.5; placental
defects included significantly reduced labyrinthine branching
morphogenesis, widespread penetration of the labyrinth by
spongiotrophoblasts and abnormal distribution of trophoblast giant
cells; in heart underdevelopment of the trabeculae, remarkably
thinner myocardium, and incomplete formation of the
interventricular septum; increased HIF-α in the placenta and the
embryo proper but not in the heart; VEGF unchanged in heart and
placentae; increased expression of MASH2 and decreased
expression of TFEB and GCM1
EIIa (adenovirus promoter) General deleter,
early embryo
PHD1−/− and PHD3−/−:
Viable; normal placenta and cardiovascular development; HIF-α
and VEGF protein levels unchanged
Takeda et al.134 ROSA26-tamoxifen-
inducible ER-Cre
Mosaic PHD2−/−:
At 6 weeks, after tamoxifen treatment, ear, trachea, liver, lung,
renal cortex and brain with increased angiogenesis and
angiectasia; glomerulomegaly with capillary dilatations; severe
polycythemia; serum VEGF levels increased but no localized
VEGF mRNA upregulation
ROSA26-tamoxifen-
inducible ER-Cre
Mosaic PHD1−/− and PHD3−/−:
No significant vascular phenotype; serum VEGF levels not
increased

A summary of mouse phenotypes resulting from the conditional activation of non-degradable forms of HIF-1α (HIF1dPA) and/or HIF-2α (HIF2dPA) using Cre-loxP-mediated recombination (a), and from conditional inactivation of PHD1, 2 and 3 (b) is presented

Concluding Remarks

The pVHL E3-ubiquitin ligase complex is essential for HIF-α proteolysis under normoxia and thus plays a critical role during embryonic development and for normal tissue physiology in the adult. In this review, we have summarized findings from the analysis of tissue-specific VHL knockout mice and have discussed the role of individual HIF transcription factors in the development of VHL phenotypes and in the context of VHL-associated tumorigenesis. While pVHL has biological functions that do not involve the HIF pathway, most organ pathologies in VHL knockout mice result from constitutive activation of HIF signaling with context- and tissue-specific contributions of HIF-1 and HIF-2 transcription factors. For a more comprehensive understanding of pVHL’s HIF-independent functions in tumor suppression, development and normal tissue physiology, additional genetic studies are needed that investigate and directly compare HIF signaling in wild-type and VHL-deficient backgrounds.

Acknowledgements

VHH is supported by grants from the National Cancer Institute (NCI), the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and the American Heart Association (AHA). Because of space limitations, the authors were unable to accommodate all relevant references and apologize to our colleagues whose work was not cited.

Abbreviations

ARNT

arylhydrocarbon receptor nuclear translocator

CC-RCC

renal cell carcinoma of the clear cell type

Cre

Cre recombinase

EPO

erythropoietin

HIF

hypoxia-inducible factor

PEPCK

phosphoenolpyruvate carboxykinase

PHD protein

prolyl-hydroxylase domain protein

pVHL

von Hippel–Lindau protein

VEGF

vascular endothelial growth factor

VHL

von Hippel–Lindau

THP

Tamm–Horsfall protein

References

  • 1.Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, et al. von Hippel– Lindau disease. Lancet. 2003;361:2059–2067. doi: 10.1016/S0140-6736(03)13643-4. [DOI] [PubMed] [Google Scholar]
  • 2.Gnarra JR, Tory K, Weng Y, Schmidt L, Wei MH, Li H, et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet. 1994;7:85–90. doi: 10.1038/ng0594-85. [DOI] [PubMed] [Google Scholar]
  • 3.Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, et al. Identification of the von Hippel–Lindau disease tumor suppressor gene [see comments] Science. 1993;260:1317–1320. doi: 10.1126/science.8493574. [DOI] [PubMed] [Google Scholar]
  • 4.Iliopoulos O, Levy AP, Jiang C, Kaelin WG, Jr, Goldberg MA. Negative regulation of hypoxia-inducible genes by the von Hippel–Lindau protein. Proc Natl Acad Sci USA. 1996;93:10595–10599. doi: 10.1073/pnas.93.20.10595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gnarra JR, Zhou S, Merrill MJ, Wagner JR, Krumm A, Papavassiliou E, et al. Post-transcriptional regulation of vascular endothelial growth factor mRNA by the product of the VHL tumor suppressor gene. Proc Natl Acad Sci USA. 1996;93:10589–10594. doi: 10.1073/pnas.93.20.10589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Siemeister G, Weindel K, Mohrs K, Barleon B, Martiny-Baron G, Marme D. Reversion of deregulated expression of vascular endothelial growth factor in human renal carcinoma cells by von Hippel–Lindau tumor suppressor protein. Cancer Res. 1996;56:2299–2301. [PubMed] [Google Scholar]
  • 7.Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis [see comments] Nature. 1999;399:271–275. doi: 10.1038/20459. [DOI] [PubMed] [Google Scholar]
  • 8.Kibel A, Iliopoulos O, DeCaprio JA, Kaelin WG., Jr Binding of the von Hippel–Lindau tumor suppressor protein to Elongin B and C [see comments] Science. 1995;269:1444–1446. doi: 10.1126/science.7660130. [DOI] [PubMed] [Google Scholar]
  • 9.Pause A, Lee S, Worrell RA, Chen DY, Burgess WH, Linehan WM, et al. The von Hippel–Lindau tumor-suppressor gene product forms a stable complex with human CUL-2, a member of the Cdc53 family of proteins. Proc Natl Acad Sci USA. 1997;94:2156–2161. doi: 10.1073/pnas.94.6.2156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kamura T, Koepp DM, Conrad MN, Skowyra D, Moreland RJ, Iliopoulos O, et al. Rbx1, a component of the VHL tumor suppressor complex and SCF ubiquitin ligase [see comments] Science. 1999;284:657–661. doi: 10.1126/science.284.5414.657. [DOI] [PubMed] [Google Scholar]
  • 11.Stebbins CE, Kaelin WG, Jr, Pavletich NP. Structure of the VHL-ElonginC-ElonginB complex: implications for VHL tumor suppressor function. Science. 1999;284:455–461. doi: 10.1126/science.284.5413.455. [DOI] [PubMed] [Google Scholar]
  • 12.Feldman DE, Thulasiraman V, Ferreyra RG, Frydman J. Formation of the VHL-elongin BC tumor suppressor complex is mediated by the chaperonin TRiC. Mol Cell. 1999;4:1051–1061. doi: 10.1016/s1097-2765(00)80233-6. [DOI] [PubMed] [Google Scholar]
  • 13.Iwai K, Yamanaka K, Kamura T, Minato N, Conaway RC, Conaway JW, et al. Identification of the von Hippel–lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proc Natl Acad Sci USA. 1999;96:12436–12441. doi: 10.1073/pnas.96.22.12436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lisztwan J, Imbert G, Wirbelauer C, Gstaiger M, Krek W. The von Hippel–Lindau tumor suppressor protein is a component of an E3 ubiquitin-protein ligase activity. Genes Dev. 1999;13:1822–1833. doi: 10.1101/gad.13.14.1822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Salceda S, Caro J. Hypoxia-inducible factor 1alpha (HIF-1alpha) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions Its stabilization by hypoxia depends on redox-induced changes. J Biol Chem. 1997;272:22642–22647. doi: 10.1074/jbc.272.36.22642. [DOI] [PubMed] [Google Scholar]
  • 16.Cockman ME, Masson N, Mole DR, Jaakkola P, Chang GW, Clifford SC, et al. Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel–Lindau tumor suppressor protein. J Biol Chem. 2000;275:25733–25741. doi: 10.1074/jbc.M002740200. [DOI] [PubMed] [Google Scholar]
  • 17.Tanimoto K, Makino Y, Pereira T, Poellinger L. Mechanism of regulation of the hypoxia-inducible factor-1 alpha by the von Hippel–Lindau tumor suppressor protein. EMBO J. 2000;19:4298–4309. doi: 10.1093/emboj/19.16.4298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, et al. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel–Lindau protein. Nat Cell Biol. 2000;2:423–427. doi: 10.1038/35017054. [DOI] [PubMed] [Google Scholar]
  • 19.Kamura T, Sato S, Iwai K, Czyzyk-Krzeska M, Conaway RC, Conaway JW. Activation of HIF1alpha ubiquitination by a reconstituted von Hippel–Lindau (VHL) tumor suppressor complex. Proc Natl Acad Sci USA. 2000;97:10430–10435. doi: 10.1073/pnas.190332597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Maynard MA, Qi H, Chung J, Lee EH, Kondo Y, Hara S, et al. Multiple splice variants of the human HIF-3alpha locus are targets of the VHL E3 ubiquitin ligase complex. J Biol Chem. 2003;278:1032–1040. doi: 10.1074/jbc.M208681200. [DOI] [PubMed] [Google Scholar]
  • 21.Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, et al. Targeting of HIF-alpha to the von Hippel–Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science. 2001;292:468–472. doi: 10.1126/science.1059796. [DOI] [PubMed] [Google Scholar]
  • 22.Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, et al. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science. 2001;292:464–468. doi: 10.1126/science.1059817. [DOI] [PubMed] [Google Scholar]
  • 23.Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O’Rourke J, Mole DR, et al. C.elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell. 2001;107:43–54. doi: 10.1016/s0092-8674(01)00507-4. [DOI] [PubMed] [Google Scholar]
  • 24.Hon WC, Wilson MI, Harlos K, Claridge TD, Schofield CJ, Pugh CW, et al. Structural basis for the recognition of hydroxyproline in HIF-1 alpha by pVHL. Nature. 2002;417:975–978. doi: 10.1038/nature00767. [DOI] [PubMed] [Google Scholar]
  • 25.Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ. Independent function of two destruction domains in hypoxia-inducible factor-alpha chains activated by prolyl hydroxylation. EMBO J. 2001;20:5197–5206. doi: 10.1093/emboj/20.18.5197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Yu F, White SB, Zhao Q, Lee FS. HIF-1alpha binding to VHL is regulated by stimulus-sensitive proline hydroxylation. Proc Natl Acad Sci USA. 2001;98:9630–9635. doi: 10.1073/pnas.181341498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bruick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science. 2001;294:1337–1340. doi: 10.1126/science.1066373. [DOI] [PubMed] [Google Scholar]
  • 28.Wenger RH, Stiehl DP, Camenisch G. Integration of oxygen signaling at the consensus HRE. Sci STKE. 2005;2005:re12. doi: 10.1126/stke.3062005re12. [DOI] [PubMed] [Google Scholar]
  • 29.Hu CJ, Wang LY, Chodosh LA, Keith B, Simon MC. Differential roles of hypoxia-inducible factor 1alpha (HIF-1alpha) and HIF-2alpha in hypoxic gene regulation. Mol Cell Biol. 2003;23:9361–9374. doi: 10.1128/MCB.23.24.9361-9374.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Warnecke C, Zaborowska Z, Kurreck J, Erdmann VA, Frei U, Wiesener M, et al. Differentiating the functional role of hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha (EPAS-1) by the use of RNA interference: erythropoietin is a HIF-2alpha target gene in Hep3B and Kelly cells. FASEB J. 2004;18:1462–1464. doi: 10.1096/fj.04-1640fje. [DOI] [PubMed] [Google Scholar]
  • 31.Rankin EB, Higgins DF, Walisser JA, Johnson RS, Bradfield CA, Haase VH. Inactivation of the arylhydrocarbon receptor nuclear translocator (Arnt) suppresses von Hippel–Lindau disease-associated vascular tumors in mice. Mol Cell Biol. 2005;25:3163–3172. doi: 10.1128/MCB.25.8.3163-3172.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rankin EB, Biju MP, Liu Q, Unger TL, Rha J, Johnson RS, et al. Hypoxia-inducible factor-2 (HIF-2) regulates hepatic erythropoietin in vivo. J Clin Invest. 2007;117:1068–1077. doi: 10.1172/JCI30117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gruber M, Hu CJ, Johnson RS, Brown EJ, Keith B, Simon MC. Acute postnatal ablation of Hif-2alpha results in anemia. Proc Natl Acad Sci USA. 2007;104:2301–2306. doi: 10.1073/pnas.0608382104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.An WG, Kanekal M, Simon MC, Maltepe E, Blagosklonny MV, Neckers LM. Stabilization of wild-type p53 by hypoxia-inducible factor 1alpha. Nature. 1998;392:405–408. doi: 10.1038/32925. [DOI] [PubMed] [Google Scholar]
  • 35.Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, et al. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev. 2000;14:34–44. [PMC free article] [PubMed] [Google Scholar]
  • 36.Koshiji M, Kageyama Y, Pete EA, Horikawa I, Barrett JC, Huang LE. HIF-1alpha induces cell cycle arrest by functionally counteracting Myc. EMBO J. 2004;23:1949–1956. doi: 10.1038/sj.emboj.7600196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gustafsson MV, Zheng X, Pereira T, Gradin K, Jin S, Lundkvist J, et al. Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell. 2005;9:617–628. doi: 10.1016/j.devcel.2005.09.010. [DOI] [PubMed] [Google Scholar]
  • 38.Lando D, Peet DJ, Whelan DA, Gorman JJ, Whitelaw ML. Asparagine hydroxylation of the HIF transactivation domain a hypoxic switch. Science. 2002;295:858–861. doi: 10.1126/science.1068592. [DOI] [PubMed] [Google Scholar]
  • 39.Mahon PC, Hirota K, Semenza GL. FIH-1: a novel protein that interacts with HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev. 2001;15:2675–2686. doi: 10.1101/gad.924501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lando D, Peet DJ, Gorman JJ, Whelan DA, Whitelaw ML, Bruick RK. FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev. 2002;16:1466–1471. doi: 10.1101/gad.991402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Stolze IP, Tian YM, Appelhoff RJ, Turley H, Wykoff CC, Gleadle JM, et al. Genetic analysis of the role of the asparaginyl hydroxylase factor inhibiting hypoxia-inducible factor (HIF) in regulating HIF transcriptional target genes. J Biol Chem. 2004;279:42719–42725. doi: 10.1074/jbc.M406713200. [DOI] [PubMed] [Google Scholar]
  • 42.Kurban G, Duplan E, Ramlal N, Hudon V, Sado Y, Ninomiya Y, et al. Collagen matrix assembly is driven by the interaction of von Hippel–Lindau tumor suppressor protein with hydroxylated collagen IV alpha 2. Oncogene. 2007 Aug 13; doi: 10.1038/sj.onc.1210709. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 43.Kurban G, Hudon V, Duplan E, Ohh M, Pause A. Characterization of a von Hippel Lindau pathway involved in extracellular matrix remodeling, cell invasion, and angiogenesis. Cancer Res. 2006;66:1313–1319. doi: 10.1158/0008-5472.CAN-05-2560. [DOI] [PubMed] [Google Scholar]
  • 44.Ohh M, Yauch RL, Lonergan KM, Whaley JM, Stemmer-Rachamimov AO, Louis DN, et al. The von Hippel–Lindau tumor suppressor protein is required for proper assembly of an extracellular fibronectin matrix. Mol Cell. 1998;1:959–968. doi: 10.1016/s1097-2765(00)80096-9. [DOI] [PubMed] [Google Scholar]
  • 45.Koochekpour S, Jeffers M, Wang PH, Gong C, Taylor GA, Roessler LM, et al. The von Hippel–Lindau tumor suppressor gene inhibits hepatocyte growth factor/scatter factor-induced invasion and branching morphogenesis in renal carcinoma cells. Mol Cell Biol. 1999;19:5902–5912. doi: 10.1128/mcb.19.9.5902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Davidowitz EJ, Schoenfeld AR, Burk RD. VHL induces renal cell differentiation and growth arrest through integration of cell-cell and cell-extracellular matrix signaling. Mol Cell Biol. 2001;21:865–874. doi: 10.1128/MCB.21.3.865-874.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Bishop T, Lau KW, Epstein AC, Kim SK, Jiang M, O’Rourke D, et al. Genetic analysis of pathways regulated by the von Hippel–Lindau tumor suppressor in Caenorhabditis elegans. PLoS Biol. 2004;2:e289. doi: 10.1371/journal.pbio.0020289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Calzada MJ, Esteban MA, Feijoo-Cuaresma M, Castellanos MC, Naranjo-Suarez S, Temes E, et al. von Hippel–Lindau tumor suppressor protein regulates the assembly of intercellular junctions in renal cancer cells through hypoxia-inducible factor-independent mechanisms. Cancer Res. 2006;66:1553–1560. doi: 10.1158/0008-5472.CAN-05-3236. [DOI] [PubMed] [Google Scholar]
  • 49.Yang H, Minamishima YA, Yan Q, Schlisio S, Ebert BL, Zhang X, et al. pVHL acts as an adaptor to promote the inhibitory phosphorylation of the NF-kappaB agonist Card9 by CK2. Mol Cell. 2007;28:15–27. doi: 10.1016/j.molcel.2007.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Peruzzi B, Athauda G, Bottaro DP. The von Hippel–Lindau tumor suppressor gene product represses oncogenic beta-catenin signaling in renal carcinoma cells. Proc Natl Acad Sci USA. 2006;103:14531–14536. doi: 10.1073/pnas.0606850103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Roe JS, Kim H, Lee SM, Kim ST, Cho EJ, Youn HD. p53 stabilization and transactivation by a von Hippel–Lindau protein. Mol Cell. 2006;22:395–405. doi: 10.1016/j.molcel.2006.04.006. [DOI] [PubMed] [Google Scholar]
  • 52.Hergovich A, Lisztwan J, Barry R, Ballschmieter P, Krek W. Regulation of microtubule stability by the von Hippel–Lindau tumour suppressor protein pVHL. Nat Cell Biol. 2003;5:64–70. doi: 10.1038/ncb899. [DOI] [PubMed] [Google Scholar]
  • 53.Thoma CR, Frew IJ, Hoerner CR, Montani M, Moch H, Krek W. pVHL and GSK3beta are components of a primary cilium-maintenance signalling network. Nat Cell Biol. 2007;9:588–595. doi: 10.1038/ncb1579. [DOI] [PubMed] [Google Scholar]
  • 54.Schermer B, Ghenoiu C, Bartram M, Muller RU, Kotsis F, Hohne M, et al. The von Hippel–Lindau tumor suppressor protein controls ciliogenesis by orienting microtubule growth. J Cell Biol. 2006;175:547–554. doi: 10.1083/jcb.200605092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lutz MS, Burk RD. Primary cilium formation requires von Hippel–Lindau gene function in renal-derived cells. Cancer Res. 2006;66:6903–6907. doi: 10.1158/0008-5472.CAN-06-0501. [DOI] [PubMed] [Google Scholar]
  • 56.Zhou MI, Wang H, Foy RL, Ross JJ, Cohen HT. Tumor suppressor von Hippel–Lindau (VHL) stabilization of Jade-1 protein occurs through plant homeodomains and is VHL mutation dependent. Cancer Res. 2004;64:1278–1286. doi: 10.1158/0008-5472.can-03-0884. [DOI] [PubMed] [Google Scholar]
  • 57.Zhou MI, Wang H, Ross JJ, Kuzmin I, Xu C, Cohen HT. The von Hippel–Lindau tumor suppressor stabilizes novel plant homeodomain protein Jade-1. J Biol Chem. 2002;277:39887–39898. doi: 10.1074/jbc.M205040200. [DOI] [PubMed] [Google Scholar]
  • 58.Pal S, Claffey KP, Dvorak HF, Mukhopadhyay D. The von Hippel–Lindau gene product inhibits vascular permeability factor/vascular endothelial growth factor expression in renal cell carcinoma by blocking protein kinase C pathways. J Biol Chem. 1997;272:27509–27512. doi: 10.1074/jbc.272.44.27509. [DOI] [PubMed] [Google Scholar]
  • 59.Pal S, Claffey KP, Cohen HT, Mukhopadhyay D. Activation of Sp1-mediated vascular permeability factor/vascular endothelial growth factor transcription requires specific interaction with protein kinase C zeta. J Biol Chem. 1998;273:26277–26280. doi: 10.1074/jbc.273.41.26277. [DOI] [PubMed] [Google Scholar]
  • 60.Datta K, Nambudripad R, Pal S, Zhou M, Cohen HT, Mukhopadhyay D. Inhibition of insulin-like growth factor-I-mediated cell signaling by the von Hippel–Lindau gene product in renal cancer. J Biol Chem. 2000;275:20700–20706. doi: 10.1074/jbc.M909970199. [DOI] [PubMed] [Google Scholar]
  • 61.Datta K, Sundberg C, Karumanchi SA, Mukhopadhyay D. The 104-123 amino acid sequence of the beta-domain of von Hippel–Lindau gene product is sufficient to inhibit renal tumor growth and invasion. Cancer Res. 2001;61:1768–1775. [PubMed] [Google Scholar]
  • 62.Okuda H, Saitoh K, Hirai S, Iwai K, Takaki Y, Baba M, et al. The von Hippel–Lindau tumor suppressor protein mediates ubiquitination of activated atypical protein kinase C. J Biol Chem. 2001;276:43611–43617. doi: 10.1074/jbc.M107880200. [DOI] [PubMed] [Google Scholar]
  • 63.Li Z, Wang D, Na X, Schoen SR, Messing EM, Wu G. The VHL protein recruits a novel KRAB-A domain protein to repress HIF-1alpha transcriptional activity. EMBO J. 2003;22:1857–1867. doi: 10.1093/emboj/cdg173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Li Z, Na X, Wang D, Schoen SR, Messing EM, Wu G. Ubiquitination of a novel deubiquitinating enzyme requires direct binding to von Hippel–Lindau tumor suppressor protein. J Biol Chem. 2002;277:4656–4662. doi: 10.1074/jbc.M108269200. [DOI] [PubMed] [Google Scholar]
  • 65.Kuznetsova AV, Meller J, Schnell PO, Nash JA, Ignacak ML, Sanchez Y, et al. von Hippel–Lindau protein binds hyperphosphorylated large subunit of RNA polymerase II through a proline hydroxylation motif and targets it for ubiquitination. Proc Natl Acad Sci USA. 2003;100:2706–2711. doi: 10.1073/pnas.0436037100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Pioli PA, Rigby WF. The von Hippel–Lindau protein interacts with heteronuclear ribonucleoprotein a2 and regulates its expression. J Biol Chem. 2001;276:40346–40352. doi: 10.1074/jbc.M105391200. [DOI] [PubMed] [Google Scholar]
  • 67.Gnarra JR, Ward JM, Porter FD, Wagner JR, Devor DE, Grinberg A, et al. Defective placental vasculogenesis causes embryonic lethality in VHL-deficient mice. Proc Natl Acad Sci USA. 1997;94:9102–9107. doi: 10.1073/pnas.94.17.9102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Takeda K, Ho VC, Takeda H, Duan LJ, Nagy A, Fong GH. Placental but not heart defects are associated with elevated hypoxia-inducible factor alpha levels in mice lacking prolyl hydroxylase domain protein 2. Mol Cell Biol. 2006;26:8336–8346. doi: 10.1128/MCB.00425-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Pfander D, Kobayashi T, Knight MC, Zelzer E, Chan DA, Olsen BR, et al. Deletion of Vhlh in chondrocytes reduces cell proliferation and increases matrix deposition during growth plate development. Development. 2004;131:2497–2508. doi: 10.1242/dev.01138. [DOI] [PubMed] [Google Scholar]
  • 70.Schipani E, Ryan HE, Didrickson S, Kobayashi T, Knight M, Johnson RS. Hypoxia in cartilage: HIF-1alpha is essential for chondrocyte growth arrest and survival. Genes Dev. 2001;15:2865–2876. doi: 10.1101/gad.934301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Neumann HP, Bender BU. Genotype-phenotype correlations in von Hippel–Lindau disease. J Intern Med. 1998;243:541–545. doi: 10.1046/j.1365-2796.1998.00336.x. [DOI] [PubMed] [Google Scholar]
  • 72.Clifford SC, Cockman ME, Smallwood AC, Mole DR, Woodward ER, Maxwell PH, et al. Contrasting effects on HIF-1alpha regulation by disease-causing pVHL mutations correlate with patterns of tumourigenesis in von Hippel–Lindau disease. Hum Mol Genet. 2001;10:1029–1038. doi: 10.1093/hmg/10.10.1029. [DOI] [PubMed] [Google Scholar]
  • 73.Hoffman MA, Ohh M, Yang H, Klco JM, Ivan M, Kaelin WG., Jr von Hippel–Lindau protein mutants linked to type 2C VHL disease preserve the ability to downregulate HIF. Hum Mol Genet. 2001;10:1019–1027. doi: 10.1093/hmg/10.10.1019. [DOI] [PubMed] [Google Scholar]
  • 74.Cario H, Schwarz K, Jorch N, Kyank U, Petrides PE, Schneider DT, et al. Mutations in the von Hippel–Lindau (VHL) tumor suppressor gene and VHL-haplotype analysis in patients with presumable congenital erythrocytosis. Haematologica. 2005;90:19–24. [PubMed] [Google Scholar]
  • 75.Ang SO, Chen H, Hirota K, Gordeuk VR, Jelinek J, Guan Y, et al. Disruption of oxygen homeostasis underlies congenital Chuvash polycythemia. Nat Genet. 2002;32:614–621. doi: 10.1038/ng1019. [DOI] [PubMed] [Google Scholar]
  • 76.Haase VH, Glickman JN, Socolovsky M, Jaenisch R. Vascular tumors in livers with targeted inactivation of the von Hippel–Lindau tumor suppressor. Proc Natl Acad Sci USA. 2001;98:1583–1588. doi: 10.1073/pnas.98.4.1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rojiani AM, Owen DA, Berry K, Woodhurst B, Anderson FH, Scudamore CH, et al. Hepatic hemangioblastoma. An unusual presentation in a patient with von Hippel–Lindau disease. Am J Surg Pathol. 1991;15:81–86. [PubMed] [Google Scholar]
  • 78.McGrath FP, Gibney RG, Morris DC, Owen DA, Erb SR. Case report: multiple hepatic and pulmonary haemangioblastomas - a new manifestation of von Hippel–Lindau disease. Clin Radiol. 1992;45:37–39. doi: 10.1016/s0009-9260(05)81467-9. [DOI] [PubMed] [Google Scholar]
  • 79.Ma W, Tessarollo L, Hong SB, Baba M, Southon E, Back TC, et al. Hepatic vascular tumors, angiectasis in multiple organs, and impaired spermatogenesis in mice with conditional inactivation of the VHL gene. Cancer Res. 2003;63:5320–5328. [PubMed] [Google Scholar]
  • 80.Ding M, Cui S, Li C, Jothy S, Haase V, Steer BM, et al. Loss of the tumor suppressor Vhlh leads to upregulation of Cxcr4 and rapidly progressive glomerulonephritis in mice. Nat Med. 2006;12:1081–1087. doi: 10.1038/nm1460. [DOI] [PubMed] [Google Scholar]
  • 81.Brukamp K, Jim B, Moeller MJ, Haase VH. Hypoxia and podocyte-specific Vhlh deletion confer risk of glomerular disease. Am J Physiol Renal Physiol. 2007;293:1397–1407. doi: 10.1152/ajprenal.00133.2007. [DOI] [PubMed] [Google Scholar]
  • 82.Steenhard B, Isom K, Stroganova L, St John PL, Freeburg PB, Holzman LB, et al. Podocyte-selective deletion of von Hippel–Lindau (VHL) protein causes albuminuria. J Am Soc Nephrol; Philadelphia, PA. ASN Annual Meeting, 2005; 2005. p. 667A. [Google Scholar]
  • 83.Rankin EB, Tomaszewski JE, Haase VH. Renal cyst development in mice with conditional inactivation of the von Hippel–Lindau tumor suppressor. Cancer Res. 2006;66:2576–2583. doi: 10.1158/0008-5472.CAN-05-3241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Peyssonnaux C, Zinkernagel AS, Schuepbach RA, Rankin E, Vaulont S, Haase VH, et al. Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs) J Clin Invest. 2007;117:1926–1932. doi: 10.1172/JCI31370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Neumann AK, Yang J, Biju MP, Joseph SK, Johnson RS, Haase VH, et al. Hypoxia inducible factor 1 alpha regulates T cell receptor signal transduction. Proc Natl Acad Sci USA. 2005;102:17071–17076. doi: 10.1073/pnas.0506070102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Biju MP, Neumann AK, Bensinger SJ, Johnson RS, Turka LA, Haase VH. Vhlh gene deletion induces Hif-1-mediated cell death in thymocytes. Mol Cell Biol. 2004;24:9038–9047. doi: 10.1128/MCB.24.20.9038-9047.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, Mackman N, et al. HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell. 2003;112:645–657. doi: 10.1016/s0092-8674(03)00154-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Karhausen J, Furuta GT, Tomaszewski JE, Johnson RS, Colgan SP, Haase VH. Epithelial hypoxia-inducible factor-1 is protective in murine experimental colitis. J Clin Invest. 2004;114:1098–1106. doi: 10.1172/JCI21086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Seagroves TN, Liao D, Cardiff R, Haase VH, Johnson RS. Biology of Hypoxia: The Role of Oxygen Sensing in Development, Normal Function and Disease. Keystone Symposia: Steamboat Springs, Colorado; 2004. Deletion of vhl impairs normal mammary gland development; p. 100. [Google Scholar]
  • 90.Boutin A, Haase VH, Johnson RS. Biology of Hypoxia: The Role of Oxygen Sensing in Development, Normal Function and Disease. Keystone Symposia: Steamboat Springs, Colorado; 2004. Response to oxygenation in differentiation, function and pathology of the epidermis; p. 114. [Google Scholar]
  • 91.Tang N, Mack F, Haase VH, Simon MC, Johnson RS. pVHL function is essential for endothelial extracellular matrix deposition. Mol Cell Biol. 2006;26:2519–2530. doi: 10.1128/MCB.26.7.2519-2530.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wang Y, Wan C, Deng L, Liu X, Cao X, Gilbert SR, et al. The hypoxia-inducible factor alpha pathway couples angiogenesis to osteogenesis during skeletal development. J Clin Invest. 2007;117:1616–1626. doi: 10.1172/JCI31581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hong SB, Furihata M, Baba M, Zbar B, Schmidt LS. Vascular defects and liver damage by the acute inactivation of the VHL gene during mouse embryogenesis. Lab Invest. 2006;86:664–675. doi: 10.1038/labinvest.3700431. [DOI] [PubMed] [Google Scholar]
  • 94.Vortmeyer AO, Gnarra JR, Emmert-Buck MR, Katz D, Linehan WM, Oldfield EH, et al. von Hippel–Lindau gene deletion detected in the stromal cell component of a cerebellar hemangioblastoma associated with von Hippel–Lindau disease. Hum Pathol. 1997;28:540–543. doi: 10.1016/s0046-8177(97)90075-7. [DOI] [PubMed] [Google Scholar]
  • 95.Lach B, Gregor A, Rippstein P, Omulecka A. Angiogenic histogenesis of stromal cells in hemangioblastoma: ultrastructural and immunohistochemical study. Ultrastruct Pathol. 1999;23:299–310. doi: 10.1080/019131299281446. [DOI] [PubMed] [Google Scholar]
  • 96.Vortmeyer AO, Frank S, Jeong SY, Yuan K, Ikejiri B, Lee YS, et al. Developmental arrest of angioblastic lineage initiates tumorigenesis in von Hippel–Lindau disease. Cancer Res. 2003;63:7051–7055. [PubMed] [Google Scholar]
  • 97.Neumann HP, Zbar B. Renal cysts, renal cancer and von Hippel–Lindau disease [editorial] Kidney Int. 1997;51:16–26. doi: 10.1038/ki.1997.3. [DOI] [PubMed] [Google Scholar]
  • 98.Kleymenova E, Everitt JI, Pluta L, Portis M, Gnarra JR, Walker CL. Susceptibility to vascular neoplasms but no increased susceptibility to renal carcinogenesis in Vhl knockout mice. Carcinogenesis. 2004;25:309–315. doi: 10.1093/carcin/bgh017. [DOI] [PubMed] [Google Scholar]
  • 99.Da Silva JL, Lacombe C, Bruneval P, Casadevall N, Leporrier M, Camilleri JP, et al. Tumor cells are the site of erythropoietin synthesis in human renal cancers associated with polycythemia. Blood. 1990;75:577–582. [PubMed] [Google Scholar]
  • 100.Krieg M, Marti HH, Plate KH. Coexpression of erythropoietin and vascular endothelial growth factor in nervous system tumors associated with von Hippel–Lindau tumor suppressor gene loss of function. Blood. 1998;92:3388–3393. [PubMed] [Google Scholar]
  • 101.Hickey MM, Lam JC, Bezman NA, Rathmell WK, Simon MC. von Hippel–Lindau mutation in mice recapitulates Chuvash polycythemia via hypoxia-inducible factor-2alpha signaling and splenic erythropoiesis. J Clin Invest. 2007;117:3879–3889. doi: 10.1172/JCI32614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yoder BK. Role of primary cilia in the pathogenesis of polycystic kidney disease. J Am Soc Nephrol. 2007;18:1381–1388. doi: 10.1681/ASN.2006111215. [DOI] [PubMed] [Google Scholar]
  • 103.Esteban MA, Harten SK, Tran MG, Maxwell PH. Formation of primary cilia in the renal epithelium is regulated by the von Hippel–Lindau tumor suppressor protein. J Am Soc Nephrol. 2006;17:1801–1806. doi: 10.1681/ASN.2006020181. [DOI] [PubMed] [Google Scholar]
  • 104.Maher ER, Kaelin WG., Jr von Hippel–Lindau disease. Medicine (Baltimore) 1997;76:381–391. doi: 10.1097/00005792-199711000-00001. [DOI] [PubMed] [Google Scholar]
  • 105.Mandriota SJ, Turner KJ, Davies DR, Murray PG, Morgan NV, Sowter HM, et al. HIF activation identifies early lesions in VHL kidneys: evidence for site-specific tumor suppressor function in the nephron. Cancer Cell. 2002;1:459–468. doi: 10.1016/s1535-6108(02)00071-5. [DOI] [PubMed] [Google Scholar]
  • 106.Esteban MA, Tran MG, Harten SK, Hill P, Castellanos MC, Chandra A, et al. Regulation of E-cadherin expression by VHL and hypoxia-inducible factor. Cancer Res. 2006;66:3567–3575. doi: 10.1158/0008-5472.CAN-05-2670. [DOI] [PubMed] [Google Scholar]
  • 107.Evans AJ, Russell RC, Roche O, Burry TN, Fish JE, Chow VW, et al. VHL promotes E2 box-dependent E-cadherin transcription by HIF-mediated regulation of SIP1 and snail. Mol Cell Biol. 2007;27:157–169. doi: 10.1128/MCB.00892-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Krishnamachary B, Zagzag D, Nagasawa H, Rainey K, Okuyama H, Baek JH, et al. Hypoxia-inducible factor-1-dependent repression of E-cadherin in von Hippel–Lindau tumor suppressor-null renal cell carcinoma mediated by TCF3, ZFHX1A, and ZFHX1B. Cancer Res. 2006;66:2725–2731. doi: 10.1158/0008-5472.CAN-05-3719. [DOI] [PubMed] [Google Scholar]
  • 109.Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell. 2003;3:347–361. doi: 10.1016/s1535-6108(03)00085-0. [DOI] [PubMed] [Google Scholar]
  • 110.Thomas GV, Tran C, Mellinghoff IK, Welsbie DS, Chan E, Fueger B, et al. Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer. Nat Med. 2006;12:122–127. doi: 10.1038/nm1337. [DOI] [PubMed] [Google Scholar]
  • 111.Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley EJ, Krek W. Chemokine receptor CXCR4 downregulated by von Hippel–Lindau tumour suppressor pVHL. Nature. 2003;425:307–311. doi: 10.1038/nature01874. [DOI] [PubMed] [Google Scholar]
  • 112.Gervais ML, Henry PC, Saravanan A, Burry TN, Gallie BL, Jewett MA, et al. Nuclear E-cadherin and VHL immunoreactivity are prognostic indicators of clear-cell renal cell carcinoma. Lab Invest. 2007;87:1252–1264. doi: 10.1038/labinvest.3700684. [DOI] [PubMed] [Google Scholar]
  • 113.Turner KJ, Moore JW, Jones A, Taylor CF, Cuthbert-Heavens D, Han C, et al. Expression of hypoxia-inducible factors in human renal cancer: relationship to angiogenesis and to the von Hippel–Lindau gene mutation. Cancer Res. 2002;62:2957–2961. [PubMed] [Google Scholar]
  • 114.Rosenberger C, Mandriota S, Jurgensen JS, Wiesener MS, Horstrup JH, Frei U, et al. Expression of hypoxia-inducible factor-1alpha and −2alpha in hypoxic and ischemic rat kidneys. J Am Soc Nephrol. 2002;13:1721–1732. doi: 10.1097/01.asn.0000017223.49823.2a. [DOI] [PubMed] [Google Scholar]
  • 115.Kondo K, Klco JM, Nakamura E, Lechpammer M, Kaelin WG. Inhibition of HIF is necessary for tumor suppression by the von Hippel–Lindau protein. Cancer Cell. 2002;1:237–246. doi: 10.1016/s1535-6108(02)00043-0. [DOI] [PubMed] [Google Scholar]
  • 116.Maranchi JK, Vasselli JR, Riss J, Bonifacio JS, Linehan WM, Klausner RD. The contribution of VHL subtrate binding and HIF-1ato the phenotype of vhl loss in renal cell carcinoma. Cancer Cell. 2002;1:247–253. doi: 10.1016/s1535-6108(02)00044-2. [DOI] [PubMed] [Google Scholar]
  • 117.Zimmer M, Doucette D, Siddiqui N, Iliopoulos O. Inhibition of hypoxia-inducible factor is sufficient for growth suppression of VHL−/− tumors. Mol Cancer Res. 2004;2:89–95. [PubMed] [Google Scholar]
  • 118.Kondo K, Kim WY, Lechpammer M, Kaelin WG., Jr Inhibition of HIF2alpha is sufficient to suppress pVHL-defective tumor growth. PLoS Biol. 2003;1:E83. doi: 10.1371/journal.pbio.0000083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Smith K, Gunaratnam L, Morley M, Franovic A, Mekhail K, Lee S. Silencing of epidermal growth factor receptor suppresses hypoxia-inducible factor-2-driven VHL−/− renal cancer. Cancer Res. 2005;65:5221–5230. doi: 10.1158/0008-5472.CAN-05-0169. [DOI] [PubMed] [Google Scholar]
  • 120.Raval RR, Lau KW, Tran MG, Sowter HM, Mandriota SJ, Li JL, et al. Contrasting properties of hypoxia-inducible factor 1 (HIF-1) and HIF-2 in von Hippel–Lindau-associated renal cell carcinoma. Mol Cell Biol. 2005;25:5675–5686. doi: 10.1128/MCB.25.13.5675-5686.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bindra RS, Vasselli JR, Stearman R, Linehan WM, Klausner RD. VHL-mediated hypoxia regulation of cyclin D1 in renal carcinoma cells. Cancer Res. 2002;62:3014–3019. [PubMed] [Google Scholar]
  • 122.Zatyka M, da Silva NF, Clifford SC, Morris MR, Wiesener MS, Eckardt KU, et al. Identification of cyclin D1 and other novel targets for the von Hippel–Lindau tumor suppressor gene by expression array analysis and investigation of cyclin D1 genotype as a modifier in von Hippel–Lindau disease. Cancer Res. 2002;62:3803–3811. [PubMed] [Google Scholar]
  • 123.Wykoff CC, Sotiriou C, Cockman ME, Ratcliffe PJ, Maxwell P, Liu E, et al. Gene array of VHL mutation and hypoxia shows novel hypoxia-induced genes and that cyclin D1 is a VHL target gene. Br J Cancer. 2004;90:1235–1243. doi: 10.1038/sj.bjc.6601657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Gordan JD, Bertout JA, Hu CJ, Diehl JA, Simon MC. HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. Cancer Cell. 2007;11:335–347. doi: 10.1016/j.ccr.2007.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Acker T, Diez-Juan A, Aragones J, Tjwa M, Brusselmans K, Moons L, et al. Genetic evidence for a tumor suppressor role of HIF-2alpha. Cancer Cell. 2005;8:131–141. doi: 10.1016/j.ccr.2005.07.003. [DOI] [PubMed] [Google Scholar]
  • 126.Maynard MA, Evans AJ, Shi W, Kim WY, Liu FF, Ohh M. Dominant-negative HIF-3alpha4 suppresses VHL-null renal cell carcinoma progression. Cell Cycle. 2007;6:2810–2816. doi: 10.4161/cc.6.22.4947. [DOI] [PubMed] [Google Scholar]
  • 127.Pollard PJ, Spencer-Dene B, Shukla D, Howarth K, Nye E, El-Bahrawy M, et al. Targeted inactivation of fh1 causes proliferative renal cyst development and activation of the hypoxia pathway. Cancer Cell. 2007;11:311–319. doi: 10.1016/j.ccr.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 128.Mack FA, Rathmell WK, Arsham AM, Gnarra J, Keith B, Simon MC. Loss of pVHL is sufficient to cause HIF dysregulation in primary cells but does not promote tumor growth. Cancer Cell. 2003;3:75–88. doi: 10.1016/s1535-6108(02)00240-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Carmeliet P, Dor Y, Herbert JM, Fukumura D, Brusselmans K, Dewerchin M, et al. Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature. 1998;394:485–490. doi: 10.1038/28867. [DOI] [PubMed] [Google Scholar]
  • 130.Rathmell WK, Hickey MM, Bezman NA, Chmielecki CA, Carraway NC, Simon MC. In vitro and in vivo models analyzing von Hippel–Lindau disease-specific mutations. Cancer Res. 2004;64:8595–8603. doi: 10.1158/0008-5472.CAN-04-1430. [DOI] [PubMed] [Google Scholar]
  • 131.Hu CJ, Iyer S, Sataur A, Covello KL, Chodosh LA, Simon MC. Differential regulation of the transcriptional activities of hypoxia-inducible factor 1 alpha (HIF-1alpha) and HIF-2alpha in stem cells. Mol Cell Biol. 2006;26:3514–3526. doi: 10.1128/MCB.26.9.3514-3526.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Flamme I, Krieg M, Plate KH. Up-regulation of vascular endothelial growth factor in stromal cells of hemangioblastomas is correlated with up-regulation of the transcription factor HRF/HIF-2alpha. Am J Pathol. 1998;153:25–29. doi: 10.1016/s0002-9440(10)65541-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kim WY, Safran M, Buckley MR, Ebert BL, Glickman J, Bosenberg M, et al. Failure to prolyl hydroxylate hypoxia-inducible factor alpha phenocopies VHL inactivation in vivo. EMBO J. 2006;25:4650–4662. doi: 10.1038/sj.emboj.7601300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Takeda K, Cowan A, Fong GH. Essential role for prolyl hydroxylase domain protein 2 in oxygen homeostasis of the adult vascular system. Circulation. 2007;116:774–781. doi: 10.1161/CIRCULATIONAHA.107.701516. [DOI] [PubMed] [Google Scholar]
  • 135.Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J. HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1alpha in normoxia. EMBO J. 2003;22:4082–4090. doi: 10.1093/emboj/cdg392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Appelhoff RJ, Tian YM, Raval RR, Turley H, Harris AL, Pugh CW, et al. Differential function of the prolyl hydroxylases PHD1, PHD2, and PHD3 in the regulation of hypoxia-inducible factor. J Biol Chem. 2004;279:38458–38465. doi: 10.1074/jbc.M406026200. [DOI] [PubMed] [Google Scholar]

RESOURCES