401
|
Allosteric effects in the marginally stable von Hippel-Lindau tumor suppressor protein and allostery-based rescue mutant design. Proc Natl Acad Sci U S A 2008; 105:901-6. [PMID: 18195360 DOI: 10.1073/pnas.0707401105] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many multifunctional tumor suppressor proteins have low stability, a property linked to cancer development. The von Hippel-Lindau tumor suppressor protein (pVHL) is one of these proteins. pVHL forms part of the E3 ubiquitin ligase complex that regulates the degradation of the hypoxia-inducible factor (HIF). Under native conditions, free pVHL is a molten globule, but it is stabilized in the E3 complex. By using molecular dynamics simulations, we observed that the interface between the two pVHL domains is the least stable region in unbound pVHL. We designed five stable mutants: one with a mutation at the interdomain interface and the others in the alpha- or beta-domains. Experimentally, type 2B pVHL disease mutant Y98N at the HIF binding site was shown to destabilize pVHL and decrease its binding affinity to HIF. Our simulations showed that the decrease in pVHL stability and binding affinity are allosterically regulated. The mutations designed to stabilize unbound wild-type pVHL, which are away from the elongin C and HIF binding sites, successfully stabilized the Y98N pVHL-elongin C complex and lowered the binding free energy of pVHL with HIF. Our results indicated both the enthalpic and dynamic allosteric components between the elongin C and HIF binding sites in pVHL, in the alpha- and beta-domains, respectively, mediated by the interdomain interface and linker. Drugs mimicking the allosteric effects of these mutants may rescue pVHL function in von Hippel-Lindau disease.
Collapse
|
402
|
Singh KP, Roy D. Allelic loss and mutations in a new ETRG-1 gene are early events in diethylstilbestrol-induced renal carcinogenesis in Syrian hamsters. Gene 2008; 408:18-26. [DOI: 10.1016/j.gene.2007.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Revised: 10/14/2007] [Accepted: 10/15/2007] [Indexed: 11/28/2022]
|
403
|
Diagnostic Utility of S100P and von Hippel-Lindau Gene Product (pVHL) in Pancreatic Adenocarcinoma—With Implication of Their Roles in Early Tumorigenesis. Am J Surg Pathol 2008; 32:78-91. [DOI: 10.1097/pas.0b013e31815701d1] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
404
|
Genetic Counseling in Renal Masses. Adv Urol 2008:720840. [PMID: 19009041 PMCID: PMC2581790 DOI: 10.1155/2008/720840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Accepted: 09/09/2008] [Indexed: 12/04/2022] Open
Abstract
All urologists have faced patients suffering a renal cancer asking for the occurrence of the disease in their offspring and very often the answer to this question has not been well founded from the scientific point of view, and only in few cases a familial segregation tree is performed. The grate shift seen in the detection of small renal masses and renal cancer in the last decades will prompt us to know the indications for familial studies, which and when are necessary, and probably to refer those patients with a suspected familial syndrome to specialized oncological centers where the appropriate molecular and familial studies could be done. Use of molecular genetic testing for early identification of at-risk family members improves diagnostic certainty and would reduce costly screening procedures in at-risk members who have not inherited disease-causing mutations. This review will focus on the molecular bases of familial syndromes associated with small renal masses and the indications of familial studies in at-risk family members.
Collapse
|
405
|
Bottaro DP, Tan N, Linehan WM. Regulation of Angiogenesis by von Hippel Lindau Protein and HIF2. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
406
|
Cook KM, Schofield CJ. Therapeutic Strategies that Target the HIF System. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
407
|
Charlesworth PJS, Harris AL. Hypoxic Regulation of Angiogenesis by HIF-1. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
408
|
Wu KL, Miao H, Khan S. JAK kinases promote invasiveness in VHL-mediated renal cell carcinoma by a suppressor of cytokine signaling-regulated, HIF-independent mechanism. Am J Physiol Renal Physiol 2007; 293:F1836-46. [PMID: 17898043 DOI: 10.1152/ajprenal.00096.2007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
von Hippel-Lindau (VHL) disease is a cancer syndrome, which includes renal cell carcinoma (RCC), and is caused by VHL mutations. Most, but not all VHL phenotypes are due to failure of mutant VHL to regulate constitutive proteolysis of hypoxia-inducible factors (HIFs). Janus kinases (JAK1, 2, 3, and TYK2) promote cell survival and proliferation, processes tightly controlled by SOCS proteins, which have sequence and structural homology to VHL. We hypothesized that in VHL disease, RCC pathogenesis results from enhanced SOCS1 degradation, leading to upregulated JAK activity. We find that baseline JAK2, JAK3, and TYK2 activities are increased in RCC cell lines, even after serum deprivation or coincubation with cytokine inhibitors. Furthermore, JAK activity is sustained in RCC stably expressing HIF2α shRNA. Invasion through Matrigel and migration in wound-healing assays, in vitro correlates of metastasis, are significantly greater in VHL mutant RCC compared with wild-type cells, and blocked by dominant-negative JAK expression or JAK inhibitors. Finally, we observe enhanced SOCS2/SOCS1 coprecipitation and reduced SOCS1 expression due to proteasomal degradation in VHL-null RCC compared with wild-type cells. The data support a new HIF-independent mechanism of RCC metastasis, whereby SOCS2 recruits SOCS1 for ubiquitination and proteasome degradation, which lead to unrestricted JAK-dependent RCC invasion. In addition to commonly proposed RCC treatment strategies that target HIFs, our data suggest that JAK inhibition represents an alternative therapeutic approach.
Collapse
Affiliation(s)
- Karen L Wu
- Case Western Reserve Univ., School of Medicine, Dept. of Nutrition, Research Tower, RT600, 2109 Adelbert Rd., Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
409
|
Gervais ML, Henry PC, Saravanan A, Burry TN, Gallie BL, Jewett MAS, Hill RP, Evans AJ, Ohh M. Nuclear E-cadherin and VHL immunoreactivity are prognostic indicators of clear-cell renal cell carcinoma. J Transl Med 2007; 87:1252-64. [PMID: 17906660 DOI: 10.1038/labinvest.3700684] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The loss of functional von Hippel-Lindau (VHL) tumor suppressor gene is associated with the development of clear-cell renal cell carcinoma (CC-RCC). Recently, VHL was shown to promote the transcription of E-cadherin, an adhesion molecule whose expression is inversely correlated with the aggressive phenotype of numerous epithelial cancers. Here, we performed immunohistochemistry on CC-RCC tissue microarrays to determine the prognostic value of E-cadherin and VHL with respect to Fuhrman grade and clinical prognosis. Low Fuhrman grade and good prognosis associated with positive VHL and E-cadherin immunoreactivity, whereas poor prognosis and high-grade tumors associated with a lack of E-cadherin and lower frequency of VHL staining. A significant portion of CC-RCC with positive VHL immunostaining correlated with nuclear localization of C-terminally cleaved E-cadherin. DNA sequencing revealed in a majority of nuclear E-cadherin-positive CC-RCC, subtle point mutations, deletions and insertions in VHL. Furthermore, nuclear E-cadherin was not observed in chromophobe or papillary RCC, as well as matched normal kidney tissue. In addition, nuclear E-cadherin localization was recapitulated in CC-RCC xenografts devoid of functional VHL or reconstituted with synthetic mutant VHL grown in SCID mice. These findings provide the first evidence of aberrant nuclear localization of E-cadherin in CC-RCC harboring VHL mutations, and suggest potential prognostic value of VHL and E-cadherin in CC-RCC.
Collapse
Affiliation(s)
- Michelle L Gervais
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
410
|
Corgna E, Betti M, Gatta G, Roila F, De Mulder PHM. Renal cancer. Crit Rev Oncol Hematol 2007; 64:247-62. [PMID: 17662611 DOI: 10.1016/j.critrevonc.2007.04.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Revised: 04/11/2007] [Accepted: 04/11/2007] [Indexed: 11/16/2022] Open
Abstract
In Europe, renal cancer (that is neoplasia of the kidney, renal pelvis or ureter (ICD-9 189 and ICD-10 C64-C66)) ranks as the seventh most common malignancy in men amongst whom there are 29,600 new cases each year (3.5% of all cancers). Tobacco, obesity and a diet poor in vegetables are all acknowledged risk factors, along with specific occupational and environmental factors. A familial history of renal carcinoma is also likely to increase the risk. Renal carcinoma may remain clinically occult for most of its course. The classic presentation of pain, haematuria, and flank mass occurs in only 9% of patients and is often indicative of advanced disease. Approximately 30% of patients with renal carcinoma present with metastatic disease, 25% with locally advanced renal carcinoma and 45% with localized disease. Metastases are typically found in the lung, soft tissue, bone, liver, cutaneous sites, and central nervous system. The most important staging technique is a computed tomography (CT) scan of the whole abdomen. Survival rates are more favourable for patients with tumours confined to the kidney. Five-year survival for patients with metastatic renal carcinoma is comprised between 0 and 20%. Radical nephrectomy is the standard intervention for renal cancer. Intrinsic resistance to chemotherapy has long been a hallmark of renal carcinoma. Limited options are available for the systemic therapy, and no chemotherapeutic regimen is accepted as a standard of care. Biologic agents represent the major effective therapies for widespread metastatic renal cancer. An antiangiogenic strategy, the neutralization of VEGF, can slow the growth rate of advanced cancer.
Collapse
|
411
|
Abstract
Renal cell carcinoma (RCC) accounts for approximately 2.6% of all cancers in the United States. While early stage disease is curable by surgery, the median survival of metastatic disease is only 13 months. In the last decade, there has been considerable progress in understanding the genetics of RCC. The VHL tumor suppressor gene is inactivated in the majority of RCC cases. The VHL protein (pVHL) acts as an E3 ligase that targets HIF-1, the hypoxia inducible transcription factor, for degradation by the ubiquitin proteasome system (UPS). In RCC cases with mutant pVHL, HIF-1 is stabilized and aberrantly expressed in normoxia, leading to the activation of pro-survival genes such as vascular endothelial growth factor (VEGF). This review will focus on the defect in the UPS that underlies RCC and describe the development of novel therapies that target the UPS. Publication history: Republished from Current BioData's Targeted Proteins database (TPdb; ).
Collapse
Affiliation(s)
- Paul G Corn
- MD Anderson Cancer Center, GU Medical Oncology, Box 1374, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
412
|
Madsen L, Schulze A, Seeger M, Hartmann-Petersen R. Ubiquitin domain proteins in disease. BMC BIOCHEMISTRY 2007; 8 Suppl 1:S1. [PMID: 18047733 PMCID: PMC2106360 DOI: 10.1186/1471-2091-8-s1-s1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The human genome encodes several ubiquitin-like (UBL) domain proteins (UDPs). Members of this protein family are involved in a variety of cellular functions and many are connected to the ubiquitin proteasome system, an essential pathway for protein degradation in eukaryotic cells. Despite their structural similarity, the UBL domains appear to have a range of different targets, resulting in a considerable diversity with respect to UDP function. Here, we give a short summary of the biochemical and physiological roles of the UDPs, which have been linked to human diseases including neurodegeneration and cancer. Publication history: Republished from Current BioData's Targeted Proteins database (TPdb; ).
Collapse
Affiliation(s)
- Louise Madsen
- Insitute of Molecular Biology and Physiology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen, Denmark
| | | | | | | |
Collapse
|
413
|
Rodriguez AR, Fishman MN. Growing opportunities for adjuvant therapy of renal cell carcinoma: targeted drugs and vaccines. Expert Opin Pharmacother 2007; 8:2979-90. [DOI: 10.1517/14656566.8.17.2979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
414
|
Charbotel B, Gad S, Caïola D, Béroud C, Fevotte J, Bergeret A, Ferlicot S, Richard S. Trichloroethylene exposure and somatic mutations of the VHL gene in patients with Renal Cell Carcinoma. J Occup Med Toxicol 2007; 2:13. [PMID: 17997830 PMCID: PMC2211482 DOI: 10.1186/1745-6673-2-13] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 11/12/2007] [Indexed: 12/03/2022] Open
Abstract
Background We investigated the association between exposure to trichloroethylene (TCE) and mutations in the von Hippel-Lindau (VHL) gene and the subsequent risk for renal cell carcinoma (RCC). Methods Cases were recruited from a case-control study previously carried out in France that suggested an association between exposures to high levels of TCE and increased risk of RCC. From 87 cases of RCC recruited for the epidemiological study, 69 were included in the present study. All samples were evaluated by a pathologist in order to identify the histological subtype and then be able to focus on clear cell RCC. The majority of the tumour samples were fixed either in formalin or Bouin's solutions. The majority of the tumours were of the clear cell RCC subtype (48 including 2 cystic RCC). Mutation screening of the 3 VHL coding exons was carried out. A descriptive analysis was performed to compare exposed and non exposed cases of clear cell RCC in terms of prevalence of mutations in both groups. Results In the 48 cases of RCC, four VHL mutations were detected: within exon 1 (c.332G>A, p.Ser111Asn), at the exon 2 splice site (c.463+1G>C and c.463+2T>C) and within exon 3 (c.506T>C, p.Leu169Pro). No difference was observed regarding the frequency of mutations in exposed versus unexposed groups: among the clear cell RCC, 25 had been exposed to TCE and 23 had no history of occupational exposure to TCE. Two patients with a mutation were identified in each group. Conclusion This study does not confirm the association between the number and type of VHL gene mutations and exposure to TCE previously described.
Collapse
Affiliation(s)
- Barbara Charbotel
- UMRESTTE, Université Lyon 1, Université de Lyon, Domaine Rockefeller, Lyon, F-69373, France.,Hospices Civils de Lyon, Service des maladies professionnelles, Centre Hospitalier Lyon Sud, F-69495 Pierre Bénite, France
| | - Sophie Gad
- Laboratoire de Génétique Oncologique EPHE, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre 94275 Le Kremlin-Bicêtre Cedex, France.,CNRS FRE-2939, Institut de Cancérologie Gustave Roussy (IGR), 94805 Villejuif, France
| | - Delphine Caïola
- Laboratoire de Génétique Oncologique EPHE, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre 94275 Le Kremlin-Bicêtre Cedex, France.,CNRS FRE-2939, Institut de Cancérologie Gustave Roussy (IGR), 94805 Villejuif, France
| | - Christophe Béroud
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Institut Universitaire de Recherche Clinique (IURC), INSERM, U 827, Montpellier, F-34000 France
| | - Joelle Fevotte
- UMRESTTE, Université Lyon 1, Université de Lyon, Domaine Rockefeller, Lyon, F-69373, France
| | - Alain Bergeret
- UMRESTTE, Université Lyon 1, Université de Lyon, Domaine Rockefeller, Lyon, F-69373, France.,Hospices Civils de Lyon, Service des maladies professionnelles, Centre Hospitalier Lyon Sud, F-69495 Pierre Bénite, France
| | - Sophie Ferlicot
- Laboratoire d'Anatomie Pathologique, CHU de Bicêtre, 94275 Le Kremlin-Bicêtre Cedex, France
| | - Stéphane Richard
- Laboratoire de Génétique Oncologique EPHE, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre 94275 Le Kremlin-Bicêtre Cedex, France.,CNRS FRE-2939, Institut de Cancérologie Gustave Roussy (IGR), 94805 Villejuif, France
| |
Collapse
|
415
|
Santarpia L, Sarlis NJ, Santarpia M, Sherman SI, Trimarchi F, Benvenga S. Mosaicism in von Hippel-Lindau disease: an event important to recognize. J Cell Mol Med 2007; 11:1408-15. [PMID: 18205710 PMCID: PMC4401302 DOI: 10.1111/j.1582-4934.2007.00122.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Accepted: 09/05/2007] [Indexed: 11/30/2022] Open
Abstract
von Hippel-Lindau disease (VHL) is an autosomal dominant, familial neoplastic disorder with variable interfamilial and intrafamilial expression. VHL is characterized by pre-disposition to development of a combination of benign and malignant tumours affecting multiple organs. We provide molecular evidence of somatic mosaicism in nearly asymptomatic man whose daughter had VHL. The mosaic subject was found to have a cyst of the kidney and an angioma of the glans penis and had had surgery for a mandibular cyst and epididymal cystadenomas. Mosaicism could provide a genetic explanation for the clinical heterogeneity and variable severity of VHL. The real incidence of mosaicism is still unclear and the identification of mosaicism has important consequences in genetic counseling of VHL patients who appear to have de novo VHL mutations and should be considered when evaluating patients with isolated VHL-related tumours. Our results strongly suggest a complete and extensive clinical examination in the parents of each patient affected by an apparently de novo VHL germline mutation. We recommend performing a mutation screening of both parents of a proband with techniques that permit detection of low percentages of mosaicism before concluding that the proband has a de novo VHL mutation.
Collapse
Affiliation(s)
- Libero Santarpia
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas, M. D. Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Sarlis
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas, M. D. Anderson Cancer Center, Houston, TX, USA
- Department of Medical Affairs - US Pharma Ops - Oncology Sanofi-Aventis Pharmaceuticals, Bridgewater, NJ, USA
| | - Mariacarmela Santarpia
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
- Department of Medical Oncology, University of Messina, Messina, Italy
| | - Steven I Sherman
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas, M. D. Anderson Cancer Center, Houston, TX, USA
| | - Francesco Trimarchi
- Department of Molecular Endocrinology, University of Messina, Messina, Italy
| | - Salvatore Benvenga
- Department of Molecular Endocrinology, University of Messina, Messina, Italy
| |
Collapse
|
416
|
Cancer-causing mutations in a novel transcription-dependent nuclear export motif of VHL abrogate oxygen-dependent degradation of hypoxia-inducible factor. Mol Cell Biol 2007; 28:302-14. [PMID: 17967880 DOI: 10.1128/mcb.01044-07] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is thought that degradation of nuclear proteins by the ubiquitylation system requires nuclear-cytoplasmic trafficking of E3 ubiquitin ligases. The von Hippel-Lindau (VHL) tumor suppressor protein is the substrate recognition component of a Cullin-2-containing E3 ubiquitin ligase that recruits hypoxia-inducible factor (HIF) for oxygen-dependent degradation. We demonstrated that VHL engages in nuclear-cytoplasmic trafficking that requires ongoing transcription to promote efficient HIF degradation. Here, we report the identification of a discreet motif, DXGX(2)DX(2)L, that directs transcription-dependent nuclear export of VHL and which is targeted by naturally occurring mutations associated with renal carcinoma and polycythemia in humans. The DXGX(2)DX(2)L motif is also found in other proteins, including poly(A)-binding protein 1, to direct its transcription-dependent nuclear export. We define DXGX(2)DX(2)L as TD-NEM (transcription-dependent nuclear export motif), since inhibition of transcription by actinomycin D or 5,6-dichlorobenzimidazole abrogates its nuclear export activity. Disease-causing mutations of key residues of TD-NEM restrain the ability of VHL to efficiently mediate oxygen-dependent degradation of HIF by altering its nuclear export dynamics without affecting interaction with its substrate. These results identify a novel nuclear export motif, further highlight the role of nuclear-cytoplasmic shuttling of E3 ligases in degradation of nuclear substrates, and provide evidence that disease-causing mutations can target subcellular trafficking.
Collapse
|
417
|
Abstract
S-nitrosothiol signaling reactions are argued to play key modulatory roles in mediating the actions of NOS in health and disease. A report by Palmer et al. in this issue of the JCI provides new insight into the in vivo biology of S-nitrosothiols (see the related article beginning on page 2592). The authors examine the chronic effects of exogenous nitrosothiol therapy and demonstrate that the commonly used antioxidant N-acetylcysteine (NAC) induces pulmonary arterial hypertension in mice. Importantly, the authors argue that the vascular pathology they observe in the lungs of these animals is functionally and morphologically equivalent to that observed in chronic hypoxia. These findings raise the concern that chronic NAC therapy may induce similar vascular pathology in patients.
Collapse
Affiliation(s)
- Philip A Marsden
- Renal Division and Department of Medicine, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
418
|
A patient with bilateral pheochromocytoma as part of a Von Hippel-Lindau (VHL) syndrome type 2C. World J Surg Oncol 2007; 5:112. [PMID: 17922902 PMCID: PMC2169240 DOI: 10.1186/1477-7819-5-112] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Accepted: 10/08/2007] [Indexed: 11/24/2022] Open
Abstract
Background Von Hippel-Lindau (VHL) disease is an autosomal dominant inherited disease. It is relatively recent that type 2C was identified as a separate group solely presenting with pheochromocytomas. As an illustration, an interesting case is presented of a pregnant woman with refractory hypertension. It proved to be the first manifestation of bilateral pheochromocytomas. The family history may indicate the diagnosis, but only identification of a germ line mutation in the DNA of a patient will confirm carriership. Case presentation A 27 year pregnant patient with intra uterine growth retardation presented with hypertension and pre-eclampsia. Magnetic resonance imaging revealed bilateral adrenal pheochromocytoma. She underwent laparoscopic adrenelectomy and a missense mutation (Gly93Ser) in exon 1 of the VHL gene on chromosome 3 (p25 – p26) was shown in the patient, her father and her daughter confirming the diagnosis of VHL. Conclusion In almost all VHL families molecular genetic analysis of DNA will demonstrate an inherited mutation. Because of the involvement in several organs, periodic clinical evaluation should take place in a well coordinated, multidisciplinary setting. VHL disease can be classified into several subtypes. VHL type 2C patients present with pheochromocytomas without evidence of haemangioblastomas in the central nervous system and/or retina and a low risk of renal cell carcinoma. Therefore, in such families, periodic clinical screening can be focussed on pheochromocytomas.
Collapse
|
419
|
Abstract
Decreased oxygen availability is a common feature during embryonic development as well of malignant tumours. Hypoxia regulates many transcription factors, and one of the most studied is the hypoxia-inducible factor (HIF). As a consequence of HIF stabilisation, the cell constitutively upregulates the hypoxic programme resulting in the expression of genes responsible for global changes in cell proliferation, angiogenesis, metastasis, invasion, de-differentiation and energy metabolism. Of the three known alpha subunits of HIF transcription factors, HIF-1alpha and HIF-2alpha have been the most studied. Their differential expression and function have been widely discussed, however no clear picture has been drawn on how these two transcription factors differently regulate common and unique target genes. Their role as oncogenes has also been suggested in several studies. In this review we provide an overview of the current knowledge on some of the most important aspects of HIFalpha regulation, its role in tumour angiogenesis and energetic metabolism. We also give an overview of how the modulation of HIF regulating pathways is a potential therapeutic target that may have benefits in the treatment of cancer.
Collapse
Affiliation(s)
- M J Calzada
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.
| | | |
Collapse
|
420
|
Abstract
Mutations of the von Hippel-Lindau (VHL) or fumarate hydratase (FH) genes lead to morphologically different renal cell carcinomas with distinct clinical courses and outcomes. The VHL protein is a part of an ubiquitin ligase complex that targets proteins for proteosomal degradation. FH is one of the mitochondrial enzymes of the Kreb's cycle. Despite two different functionalities and cellular locations, loss of either VHL or FH products has been shown to alter expression levels of hypoxia-inducible factors (HIF-1alpha and HIF-2alpha) and their downstream targets. HIF proteins are key regulators of oxygen homeostasis. Tight regulation of HIF allows for cell survival and growth at the time of hypoxic stress. HIF acts via transcriptional regulation of vascular endothelial growth factor, platelet derived growth factor, endothelial growth factor receptor, glucose transporter protein 1, erythropoietin, and transforming growth factor-alpha. Loss of VHL or FH is thought to result in a pseudohypoxic state so that cellular response pathways mediated by HIF are activated despite normal oxygen conditions. Understanding of these pseudohypoxic pathways has provided a better appreciation of the molecular mechanisms of carcinogenesis in addition to providing a rationale for targeted therapeutic approaches.
Collapse
Affiliation(s)
- Gennady Bratslavsky
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892-1107, USA
| | | | | | | |
Collapse
|
421
|
Abstract
Rapidly growing tumors invariably contain hypoxic regions. Adaptive response to hypoxia through angiogenesis, enhanced glucose metabolism and diminished but optimized mitochondrial respiration confers survival and growth advantage to hypoxic tumor cells. In this review, the roles of hypoxia, the hypoxia inducible factors, oncogenes and tumor suppressors in metabolic adaptation of tumors are discussed. These new insights into hypoxic metabolic alterations in tumors will hopefully lead us to target tumor bioenergetics for the treatment of cancers.
Collapse
Affiliation(s)
- Jung-whan Kim
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
422
|
Klein J, Zhuang Z, Lubensky I, Colby TV, Martinez F, Leslie KO. Multifocal microcysts and papillary cystadenoma of the lung in von Hippel-Lindau disease. Am J Surg Pathol 2007; 31:1292-6. [PMID: 17667557 DOI: 10.1097/pas.0b013e3180377aaf] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
von Hippel-Lindau disease is an autosomal dominant inherited disorder characterized by a predisposition to multiple neoplasms. Renal cell carcinoma and hemangioblastomas of the retina and cerebellum are the most common of these, but other neoplasms and cysts also occur throughout the body. We report a distinctive, yet never described lung lesion in a 43-year-old woman with von Hippel-Lindau disease. Molecular genetic studies confirmed the presence of a VHL gene mutation in the cells of this lesion. We discuss the salient features of this novel lesion, and hypothesize on its origin and nature.
Collapse
Affiliation(s)
- Julianne Klein
- St Boniface General Hospital, Winnipeg, Manitoba, Canada.
| | | | | | | | | | | |
Collapse
|
423
|
Rubin JS, Bottaro DP. Loss of Secreted Frizzled-Related Protein-1 Expression in Renal Cell Carcinoma Reveals a Critical Tumor Suppressor Function. Clin Cancer Res 2007; 13:4660-3. [PMID: 17699841 DOI: 10.1158/1078-0432.ccr-07-1077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jeffrey S Rubin
- Laboratory of Cellular and Molecular Biology and Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-1107, USA
| | | |
Collapse
|
424
|
Laping NJ, Everitt JI, Frazier KS, Burgert M, Portis MJ, Cadacio C, Gold LI, Walker CL. Tumor-specific efficacy of transforming growth factor-beta RI inhibition in Eker rats. Clin Cancer Res 2007; 13:3087-99. [PMID: 17505012 DOI: 10.1158/1078-0432.ccr-06-1811] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Transforming growth factor beta (TGF-beta), which generally stimulates the growth of mesenchymally derived cells but inhibits the growth of epithelial cells, has been proposed as a possible target for cancer therapy. However, concerns have been raised that whereas inhibition of TGF-beta signaling could be efficacious for lesions in which TGF-beta promotes tumor development and/or progression, systemic pharmacologic blockade of this signaling pathway could also promote the growth of epithelial lesions. EXPERIMENTAL DESIGN We examined the effect of a TGF-beta inhibitor on mesenchymal (leiomyoma) and epithelial (renal cell carcinoma) tumors in Eker rats, which are genetically predisposed to develop these tumors with a high frequency. RESULTS Blockade of TGF-beta signaling with the ALK5/type I TGF-beta R kinase inhibitor, SB-525334, was efficacious for uterine leiomyoma; significantly decreasing tumor incidence and multiplicity, and reducing the size of these mesenchymal tumors. However, SB-525334 was also mitogenic and antiapoptotic for epithelial cells in the kidney and exacerbated the growth of epithelial lesions present in the kidneys of these animals. CONCLUSION Although pharmacologic inhibition of TGF-beta signaling with SB-525334 may be efficacious for mesenchymal tumors, inhibition of this signaling pathway seems to promote the development of epithelial tumors.
Collapse
|
425
|
Kurban G, Duplan E, Ramlal N, Hudon V, Sado Y, Ninomiya Y, Pause A. Collagen matrix assembly is driven by the interaction of von Hippel-Lindau tumor suppressor protein with hydroxylated collagen IV alpha 2. Oncogene 2007; 27:1004-12. [PMID: 17700531 DOI: 10.1038/sj.onc.1210709] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene predisposes to vascular tumor formation in several organs. VHL regulates two evolutionary conserved pathways: the targeting of hydroxylated hypoxia-inducible factor-alpha (HIF-alpha) for proteasomal degradation and the remodeling of extracellular matrix (ECM). The biochemical mechanisms of the ECM assembly pathway remain poorly defined. Here, we provide evidence supporting a biochemical role for VHL in ECM assembly. We show that VHL directly binds to the collagen IV alpha 2 (COL4A2) chain and that this interaction is necessary for its assembly into the ECM. The VHL-COL4A2 interaction is dependent on endoplasmic reticulum (ER)-mediated COL4A2 hydroxylation and independent of cytosolic, hypoxia regulated HIF-alpha-modifying enzymes. We find that the N-terminal tail of COL4A2 protrudes from the ER lumen into the cytosol where it is bound by VHL. Failure of VHL to interact with COL4A2 correlates with loss of collagen IV network formation in vitro and collagen IV remodeling in vivo. Our data suggest a HIF-alpha-independent role for the VHL-COL4A2 interaction in suppression of angiogenic tumor formation through collagen IV network assembly.
Collapse
Affiliation(s)
- G Kurban
- McGill Cancer Center, Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
426
|
Wang Y, Wan C, Deng L, Liu X, Cao X, Gilbert SR, Bouxsein ML, Faugere MC, Guldberg RE, Gerstenfeld LC, Haase VH, Johnson RS, Schipani E, Clemens TL. The hypoxia-inducible factor alpha pathway couples angiogenesis to osteogenesis during skeletal development. J Clin Invest 2007; 117:1616-26. [PMID: 17549257 PMCID: PMC1878533 DOI: 10.1172/jci31581] [Citation(s) in RCA: 548] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Accepted: 03/26/2007] [Indexed: 01/14/2023] Open
Abstract
Skeletal development and turnover occur in close spatial and temporal association with angiogenesis. Osteoblasts are ideally situated in bone to sense oxygen tension and respond to hypoxia by activating the hypoxia-inducible factor alpha (HIF alpha) pathway. Here we provide evidence that HIF alpha promotes angiogenesis and osteogenesis by elevating VEGF levels in osteoblasts. Mice overexpressing HIF alpha in osteoblasts through selective deletion of the von Hippel-Lindau gene (Vhl) expressed high levels of Vegf and developed extremely dense, heavily vascularized long bones. By contrast, mice lacking Hif1a in osteoblasts had the reverse skeletal phenotype of that of the Vhl mutants: long bones were significantly thinner and less vascularized than those of controls. Loss of Vhl in osteoblasts increased endothelial sprouting from the embryonic metatarsals in vitro but had little effect on osteoblast function in the absence of blood vessels. Mice lacking both Vhl and Hif1a had a bone phenotype intermediate between those of the single mutants, suggesting overlapping functions of HIFs in bone. These studies suggest that activation of the HIF alpha pathway in developing bone increases bone modeling events through cell-nonautonomous mechanisms to coordinate the timing, direction, and degree of new blood vessel formation in bone.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Chao Wan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Lianfu Deng
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Ximeng Liu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Xuemei Cao
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Shawn R. Gilbert
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Mary L. Bouxsein
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Marie-Claude Faugere
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Robert E. Guldberg
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Louis C. Gerstenfeld
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Volker H. Haase
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Randall S. Johnson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Ernestina Schipani
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Thomas L. Clemens
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Kentucky, Lexington, Kentucky, USA.
Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
Department of Orthopedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, San Diego, California, USA.
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
Veterans Administration Medical Center, Birmingham, Alabama, USA
| |
Collapse
|
427
|
Vira MA, Novakovic KR, Pinto PA, Linehan WM. Genetic basis of kidney cancer: a model for developing molecular-targeted therapies. BJU Int 2007; 99:1223-9. [PMID: 17441915 DOI: 10.1111/j.1464-410x.2007.06814.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Manish A Vira
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1107, USA
| | | | | | | |
Collapse
|
428
|
Horii K, Suzuki Y, Kondo Y, Akimoto M, Nishimura T, Yamabe Y, Sakaue M, Sano T, Kitagawa T, Himeno S, Imura N, Hara S. Androgen-dependent gene expression of prostate-specific antigen is enhanced synergistically by hypoxia in human prostate cancer cells. Mol Cancer Res 2007; 5:383-91. [PMID: 17426252 DOI: 10.1158/1541-7786.mcr-06-0226] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The androgen receptor (AR) is implicated in prostate cancer growth, progression, and angiogenesis. Hypoxia-inducible factor-1 (HIF-1), which transcriptionally regulates hypoxia-inducible angiogenic factors, is up-regulated in prostate cancers compared with adjacent normal tissues. HIF-1 may be involved in prostate cancer as well as the AR, but the involvement of HIF-1 in prostate cancer angiogenesis and progression has not been fully elucidated. In the present study, we found that in prostate cancer LNCaP cells dihydrotestosterone enhanced the expression of GLUT-1, one of the HIF-1 target genes, and also that hypoxia enhanced the expression of prostate-specific antigen (PSA) that is one of the AR target genes and is involved in tumor invasion. Small interfering RNA that specifically inhibits HIF-1 reduced the expression levels of PSA as well as GLUT-1. Reporter gene analysis showed that dihydrotestosterone activated the HIF-1-mediated gene expression and hypoxia enhanced the AR-induced promoter activity of human PSA gene. Deletion and site-directed mutation of the 5'-flanking region of human PSA gene revealed that the sequence ACGTG between -3951 and -3947 was essential in the response to hypoxia. Furthermore, chromatin immunoprecipitation assay indicated that HIF-1 interacts with the AR on the human PSA gene promoter. These results indicated that in prostate cancers, HIF-1 might cooperate with the AR to activate the expression of several genes related to tumor angiogenesis, invasion, and progression.
Collapse
Affiliation(s)
- Kou Horii
- Department of Public Health, School of Pharmaceutical Sciences, Kitasato University, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
429
|
Abstract
Erythropoietin (EPO) is the hormonal regulator of red cell production and provided the paradigm for oxygen-regulated gene expression that led to the discovery of hypoxia-inducible factor (HIF). In this issue of the JCI, Rankin and colleagues show, using targeted gene inactivation, that induction of Epo expression in murine liver is dependent on the integrity of HIF-2alpha, and not HIF-1alpha (see the related article beginning on page 1068). These results demonstrate distinct functions for different HIF-alpha isoforms that could potentially be exploited in therapeutic approaches to anemia.
Collapse
Affiliation(s)
- Peter J Ratcliffe
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
430
|
Rankin EB, Biju MP, Liu Q, Unger TL, Rha J, Johnson RS, Simon MC, Keith B, Haase VH. Hypoxia-inducible factor-2 (HIF-2) regulates hepatic erythropoietin in vivo. J Clin Invest 2007; 117:1068-77. [PMID: 17404621 PMCID: PMC1838939 DOI: 10.1172/jci30117] [Citation(s) in RCA: 454] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2006] [Accepted: 01/09/2007] [Indexed: 01/24/2023] Open
Abstract
Erythropoiesis is critically dependent on erythropoietin (EPO), a glycoprotein hormone that is regulated by hypoxia-inducible factor (HIF). Hepatocytes are the primary source of extrarenal EPO in the adult and express HIF-1 and HIF-2, whose roles in the hypoxic induction of EPO remain controversial. In order to define the role of HIF-1 and HIF-2 in the regulation of hepatic EPO expression, we have generated mice with conditional inactivation of Hif-1alpha and/or Hif-2alpha (Epas1) in hepatocytes. We have previously shown that inactivation of the von Hippel-Lindau tumor suppressor pVHL, which targets both HIFs for proteasomal degradation, results in increased hepatic Epo production and polycythemia independent of Hif-1alpha. Here we show that conditional inactivation of Hif-2alpha in pVHL-deficient mice suppressed hepatic Epo and the development of polycythemia. Furthermore, we found that physiological Epo expression in infant livers required Hif-2alpha but not Hif-1alpha and that the hypoxic induction of liver Epo in anemic adults was Hif-2alpha dependent. Since other Hif target genes such phosphoglycerate kinase 1 (Pgk) were Hif-1alpha dependent, we provide genetic evidence that HIF-1 and HIF-2 have distinct roles in the regulation of hypoxia-inducible genes and that EPO is preferentially regulated by HIF-2 in the liver.
Collapse
Affiliation(s)
- Erinn B. Rankin
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mangatt P. Biju
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Qingdu Liu
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Travis L. Unger
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jennifer Rha
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Randall S. Johnson
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - M. Celeste Simon
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Brian Keith
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Volker H. Haase
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Division of Biological Sciences, UCSD, La Jolla, California, USA.
Abramson Family Cancer Research Institute, Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
431
|
Lehman NL, van de Rijn M, Jackson PK. Screening of tissue microarrays for ubiquitin proteasome system components in tumors. Methods Enzymol 2007; 399:334-55. [PMID: 16338367 DOI: 10.1016/s0076-6879(05)99023-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
The turnover of key proteins that mediate development, cellular proliferation, and a host of essential biological processes is controlled by the ubiquitin proteasome system (UPS). In several well-studied examples, notably in the cell cycle, regulatory proteins that control ubiquitin-dependent destruction are themselves substrates of the UPS, creating a multilayered system to ensure precise and dynamic control of protein stability. UPS regulators controlled at the level of protein stability--including the F-box protein Skp2 and the VHL protein (substrate adapter proteins for multicomponent E3 ubiquitin ligases)-- seem to be misregulated in tumors. In these cases, especially, measuring levels of critical regulatory and target proteins will often present a more biologically meaningful picture than examining relative mRNA levels, which do not always reflect corresponding protein levels. Tissue microarrays (TMAs) allow simultaneous screening of large numbers of tumors for expression of specific proteins by immunohistochemical staining of a single microscope slide prepared from a TMA paraffin block. Replicate slides prepared from the same block can be immunostained for multiple proteins functioning in a related pathway, and a semiquantitative protein expression profile for a given subset of UPS pathway components, or other subsets of proteins of interest, can be assembled. Protein expression profiles of individual tumors or tissue types can be compared and visualized by hierarchical clustering methods. These expression profiles may be used as screening tools to investigate the relative abundance of components of a biochemical pathway in tumors or other tissues. TMAs have an exciting future as tools for basic research, diagnostic pathology, and drug targeting. In this article, we provide an introduction to the use of TMAs to study the expression of UPS component proteins and substrates in tumors by immunohistochemistry.
Collapse
Affiliation(s)
- Norman L Lehman
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | | | | |
Collapse
|
432
|
Li L, Zhang L, Zhang X, Yan Q, Minamishima YA, Olumi AF, Mao M, Bartz S, Kaelin WG. Hypoxia-inducible factor linked to differential kidney cancer risk seen with type 2A and type 2B VHL mutations. Mol Cell Biol 2007; 27:5381-92. [PMID: 17526729 PMCID: PMC1952077 DOI: 10.1128/mcb.00282-07] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clear cell carcinoma of the kidney is a major cause of mortality in patients with von Hippel-Lindau (VHL) disease, which is caused by germ line mutations that inactivate the VHL tumor suppressor gene. Biallelic VHL inactivation, due to mutations or hypermethylation, is also common in sporadic clear cell renal carcinomas. The VHL gene product, pVHL, is part of a ubiquitin ligase complex that targets the alpha subunits of the heterodimeric transcription factor hypoxia-inducible factor (HIF) for destruction under well-oxygenated conditions. All VHL mutations linked to classical VHL disease compromise this pVHL function although some missense mutations result in a low risk of kidney cancer (type 2A VHL disease) while others result in a high risk (type 2B VHL disease). We found that type 2A mutants were less defective than type 2B mutants when reintroduced into VHL-/- renal carcinoma cells with respect to HIF regulation. A stabilized version of HIF2alpha promoted tumor growth by VHL-/- cells engineered to produce type 2A mutants, while knock-down of HIF2alpha in cells producing type 2B mutants had the opposite effect. Therefore, quantitative differences with respect to HIF deregulation are sufficient to account for the differential risks of kidney cancer linked to VHL mutations.
Collapse
Affiliation(s)
- Lianjie Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, 44 Binney Street, Mayer 457, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
433
|
The Importance of Pathology and Genetics for the Diagnosis and Therapy of Renal Cell Carcinoma. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/j.eursup.2007.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
434
|
Dahr SS, Cusick M, Rodriguez-Coleman H, Srivastava SK, Thompson DJ, Linehan WM, Ferris FL, Chew EY. Intravitreal anti-vascular endothelial growth factor therapy with pegaptanib for advanced von Hippel-Lindau disease of the retina. Retina 2007; 27:150-8. [PMID: 17290195 DOI: 10.1097/iae.0b013e318030a290] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This pilot study was designed to provide preliminary data concerning the safety and efficacy of pegylated anti-vascular endothelial growth factor (VEGF) therapy, pegaptanib, for patients with juxtapapillary or large peripheral angiomas secondary to von Hippel-Lindau (VHL) disease. METHODS This study was an open label, nonrandomized, prospective, pilot study of intravitreal injections of pegaptanib (3 mg/100 microL), given every 6 weeks for minimum of 6 injections. Five patients with severe ocular VHL lesions were enrolled in the study. The primary outcome of this study was a change of > or =15 letters (3 lines) in best-corrected visual acuity by 1 year. Secondary outcomes included changes in macular thickness, as determined by optical coherence tomography, and changes in fluorescein leakage. RESULTS Two of five patients completed the course of treatment and 1 year of follow-up. These two patients had progressive decrease in retinal hard exudate and reduction in central retinal thickness measured by optical coherence tomography. One of these two patients had improvement in visual acuity of 3 lines. No significant change in fluorescein leakage or tumor size was detected in either patient. Lesions in the other three patients continued to progress despite treatment, and these patients did not complete the entire treatment course. One patient developed a tractional retinal detachment. Additional serious adverse events included transient postinjection hypotony in two eyes. CONCLUSIONS Intravitreal injections of anti-VEGF therapy (pegaptanib) may decrease retinal thickening minimally and reduce retinal hard exudates in some patients with advanced VHL angiomas. This finding may be related to a reduction in vasopermeability, because there was no apparent effect of treatment on the size of the primary retinal angiomas in this small pilot study.
Collapse
Affiliation(s)
- Sam S Dahr
- Division of Epidemiology and Clinical Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
435
|
Abstract
Molecular genetic analysis of hereditary leiomyomatosis and renal cell cancer (HLRCC) unexpectedly revealed germline defects in the gene encoding the Krebs cycle enzyme fumarate hydratase (FH), stimulating great interest in the underlying mechanism of oncogenesis. It has been proposed that the associated accumulation of fumarate competitively inhibits the 2-oxoglutarate-dependent dioxygenases that regulate hypoxia-inducible factor (HIF), thus activating oncogenic hypoxia pathways. In this issue of Cancer Cell, Pollard and colleagues describe a genetic mouse model of FH deficiency that recapitulates aspects of the human disease, including HIF activation and renal cysts, enabling further insights into this unusual cancer syndrome.
Collapse
Affiliation(s)
- Peter J Ratcliffe
- The Henry Wellcome Building for Molecular Physiology, University of Oxford, Headington Campus, Roosevelt Drive, Oxford OX3 7BN, United Kingdom.
| |
Collapse
|
436
|
Tickoo SK, Alden D, Olgac S, Fine SW, Russo P, Kondagunta GV, Motzer RJ, Reuter VE. Immunohistochemical Expression of Hypoxia Inducible Factor-1α and its Downstream Molecules in Sarcomatoid Renal Cell Carcinoma. J Urol 2007; 177:1258-63. [PMID: 17382701 DOI: 10.1016/j.juro.2006.11.100] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Accepted: 11/27/2006] [Indexed: 11/26/2022]
Abstract
PURPOSE Sarcomatoid renal cell carcinomas, highly aggressive variants of renal cell carcinoma subtypes, often present with or develop metastases soon after the primary diagnosis. Most metastatic cases do not respond to immunotherapy or aggressive chemotherapy. Recently targeted therapies, particularly those targeting hypoxia inducible pathway molecules, have been tested clinically on metastatic clear cell renal cell carcinoma with promising initial results. No such studies are available on sarcomatoid renal cell carcinoma. We investigated the hypoxia inducible pathway marker immunohistochemical expression profile, and any potential therapeutic implications that such expression may have, in these tumors. MATERIALS AND METHODS Immunohistochemical staining for hypoxia inducible factor-1alpha, glucose transporter 1, carbonic anhydrase IX and vascular endothelial growth factor was performed in 22 clear cell and 12 nonclear cell sarcomatoid renal cell carcinomas. The immunoreactivity in the tumors was graded from 0 to 3+ (0-no staining, 1+-1% to 25% cells positive, 2+-26% to 50% cells positive and 3+-greater than 50% cells positive). The results were then compared with various clinical parameters to assess for associations. RESULTS Most clear cell renal cell carcinomas over expressed (2+ or 3+) hypoxia inducible factor-1alpha (in 59%), carbonic anhydrase IX (95%), glucose transporter 1 (91%) and vascular endothelial growth factor (95%). None of the nonclear cell sarcomatoid renal cell carcinomas showed 2+ or 3+ expression of hypoxia inducible factor-1alpha, carbonic anhydrase IX or glucose transporter 1, but 92% showed diffuse positivity for vascular endothelial growth factor. Over expression of carbonic anhydrase IX showed no association with survival, unlike that reported in (nonsarcomatoid) clear cell renal cell carcinoma. There was significant discordance in the staining grades among hypoxia inducible factor-1alpha, carbonic anhydrase IX and glucose transporter 1 in clear cell renal cell carcinoma, suggesting that mechanisms other than hypoxia inducible pathway may be involved in some sarcomatoid clear cell renal cell carcinoma. CONCLUSIONS Hypoxia inducible pathway markers continue to be over expressed in sarcomatoid clear cell renal cell carcinoma, and can be of diagnostic usefulness in such high grade tumors. Over expression of vascular endothelial growth factor in the clear and nonclear cell groups raises the possibility that vascular endothelial growth factor targeted therapies may have a role in the management of sarcomatoid renal cell carcinoma, and deserve further investigation.
Collapse
Affiliation(s)
- Satish K Tickoo
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA.
| | | | | | | | | | | | | | | |
Collapse
|
437
|
Sudarshan S, Pinto PA, Neckers L, Linehan WM. Mechanisms of disease: hereditary leiomyomatosis and renal cell cancer--a distinct form of hereditary kidney cancer. ACTA ACUST UNITED AC 2007; 4:104-10. [PMID: 17287871 DOI: 10.1038/ncpuro0711] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Accepted: 11/27/2006] [Indexed: 01/10/2023]
Abstract
Renal cell carcinoma (RCC) represents a group of diseases linked by their primary site of origin, the kidney. Studies of families with a genetic predisposition to the development of kidney cancer have revealed that multiple genes are involved in the molecular pathogenesis of RCC. Germline mutations in a gene that encodes a Krebs cycle enzyme have been found to result in a distinct clinical entity referred to as hereditary leiomyomatosis and renal cell cancer (HLRCC). HLRCC is inherited in an autosomal-dominant fashion. Affected individuals in HLRCC families are at risk for the development of leiomyomas of the skin and uterus as well as renal cancers. HLRCC-associated kidney tumors are often biologically aggressive. Linkage analysis has identified germline alterations in the fumarate hydratase (FH) gene associated with HLRCC. While the mechanisms of molecular carcinogenesis are not entirely understood, several lines of evidence derived from clinical and basic research suggest that pseudohypoxia might drive cellular transformation. The role of FH mutations in sporadic tumors seems to be limited. Nevertheless, continued investigation of HLRCC should provide further insight into the mechanisms of kidney cancer development, and could potentially identify targets for new therapeutic approaches to RCC.
Collapse
Affiliation(s)
- Sunil Sudarshan
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1107, USA
| | | | | | | |
Collapse
|
438
|
George DJ. Phase 2 studies of sunitinib and AG013736 in patients with cytokine-refractory renal cell carcinoma. Clin Cancer Res 2007; 13:753s-757s. [PMID: 17255305 DOI: 10.1158/1078-0432.ccr-06-2044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Frequent loss of the von Hippel-Lindau (VHL) gene product in conventional-type renal cell carcinoma results in constitutive expression of proangiogenic growth factors, including vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF). VEGF and PDGF function in a paracrine manner to stimulate tumor angiogenesis that results in a hypervascular phenotype. Dependency on this hypervascularity is underscored by the recent clinical efficacy shown by inhibition of the VEGF pathway. Most strategies that primarily target the VEGF pathway (neutralizing antibodies or receptor tyrosine kinase inhibitors) result in objective tumor responses in < or =10% of cases but show a significant delay in time to disease progression. In contrast, two multitargeted receptor tyrosine kinase inhibitors that target both VEGF and PDGF receptors (sunitinib and AG013736) have shown > or =40% objective responses with clinically important duration. Several hypotheses may explain the discrepancy of these response rates from other strategies in the class, including the synergistic effects of dual inhibition of VEGF and PDGF receptors, supported by preclinical studies. Ultimately, further clinical investigations with pharmacodynamic and correlative science end points are needed to clarify the mechanisms of action and resistance to build on the biological and clinical effects of these multitargeted agents.
Collapse
Affiliation(s)
- Daniel J George
- Department of Medicine and Surgery, Divisions of Medical Oncology and Urology, Duke University Medical Center, Durham, NC 27705, USA.
| |
Collapse
|
439
|
Lau KW, Tian YM, Raval RR, Ratcliffe PJ, Pugh CW. Target gene selectivity of hypoxia-inducible factor-alpha in renal cancer cells is conveyed by post-DNA-binding mechanisms. Br J Cancer 2007; 96:1284-92. [PMID: 17387348 PMCID: PMC2360163 DOI: 10.1038/sj.bjc.6603675] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inactivation of the von Hippel–Lindau tumour suppressor in renal cell carcinoma (RCC) leads to failure of proteolytic regulation of the α subunits of hypoxia-inducible factor (HIF), constitutive upregulation of the HIF complex, and overexpression of HIF target genes. However, recent studies have indicated that in this setting, upregulation of the closely related HIF-α isoforms, HIF-1α and HIF-2α, have contrasting effects on tumour growth, and activate distinct sets of target genes. To pursue these findings, we sought to elucidate the mechanisms underlying target gene selectivity for HIF-1α and HIF-2α. Using chromatin immunoprecipitation to probe binding to hypoxia response elements in vivo, and expression of chimaeric molecules bearing reciprocal domain exchanges between HIF-1α and HIF-2α molecules, we show that selective activation of HIF-α target gene expression is not dependent on selective DNA-binding at the target locus, but depends on non-equivalent C-terminal portions of these molecules. Our data indicate that post-DNA binding mechanisms that are dissimilar for HIF-1α and HIF-2α determine target gene selectivity in RCC cells.
Collapse
Affiliation(s)
- K W Lau
- The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Y-M Tian
- The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - R R Raval
- The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - P J Ratcliffe
- The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - C W Pugh
- The Henry Wellcome Building for Molecular Physiology, Oxford, UK
- The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Headington, Oxford, OX3 7BN, UK; E-mail:
| |
Collapse
|
440
|
Bel Aiba RS, Dimova EY, Görlach A, Kietzmann T. The role of hypoxia inducible factor-1 in cell metabolism--a possible target in cancer therapy. Expert Opin Ther Targets 2007; 10:583-99. [PMID: 16848694 DOI: 10.1517/14728222.10.4.583] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In many cancer types, intratumoural hypoxia is linked to increased expression and activity of the transcription factor hypoxia-inducible factor (HIF-1alpha), which is associated with poor patient prognosis. This increased the interest in HIF-1alpha as a cancer drug target. Further, HIF-1alpha has also a central role in the adaptive cellular programme responding to hypoxia in normal tissues. Many of the HIF-1alpha-regulated genes encode enzymes of metabolic pathways. Therefore, studying the link and the feedback mechanisms between metabolism and HIF-1alpha is of major importance to find new and specific therapeutic strategies.
Collapse
Affiliation(s)
- Rachida S Bel Aiba
- University of Kaiserslautern, Faculty of Chemistry, Department of Biochemistry, Erwin-Schrödinger Strasse 54, D-67663 Kaiserslautern, Germany
| | | | | | | |
Collapse
|
441
|
Grosfeld A, Stolze IP, Cockman ME, Pugh CW, Edelmann M, Kessler B, Bullock AN, Ratcliffe PJ, Masson N. Interaction of hydroxylated collagen IV with the von hippel-lindau tumor suppressor. J Biol Chem 2007; 282:13264-9. [PMID: 17339318 DOI: 10.1074/jbc.m611648200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The von Hippel-Lindau tumor suppressor (pVHL) targets hydroxylated alpha-subunits of hypoxia-inducible factor (HIF) for ubiquitin-mediated proteasomal destruction through direct interaction with the hydroxyproline binding pocket in its beta-domain. Although disruption of this process may contribute to VHL-associated tumor predisposition by up-regulation of HIF target genes, genetic and biochemical analyses support the existence of additional functions, including a role in the assembly of extracellular matrix. In an attempt to delineate these pathways, we searched for novel pVHL-binding proteins. Here we report a direct, hydroxylation-dependent interaction with alpha-chains of collagen IV. Interaction with pVHL was also observed with fibrillar collagen chains, but not the folded collagen triple helix. The interaction was suppressed by a wide range of tumor-associated mutations, including those that do not disturb the regulation of HIF, supporting a role in HIF-independent tumor suppressor functions.
Collapse
Affiliation(s)
- Alexandra Grosfeld
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Oxford OX3 7BN, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
442
|
Bellmunt J. Current Treatment in Advanced Renal Cell Carcinoma (RCC): Impact of Targeted Therapies in the Management of RCC. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/j.eursup.2007.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
443
|
Abstract
BACKGROUND von Hippel-Lindau Disease (VHL) is an autosomal dominant inherited systemic cancer syndrome. Recently, many advances have contributed to the understanding of VHL pathophysiology. METHODS In this article we review recent developments and summarize our findings in VHL molecular pathology related to retinal and optic nerve diseases. RESULTS Loss of heterozygosity (LOH) within the VHL gene is detected in the stromal cells surrounding the capillary endothelial cells and admixed with glial cells in ocular hemangioblastomas. This finding is in line with similar findings in VHL-associated CNS hemangioblastoma and renal clear cell carcinomas. Increases of vascular endothelial growth factor (VEGF), hypoxia induced factor (HIF), and ubiquitin are found in ocular hemangioblastomas.Interestingly, tumorlet cells, which are composed of poorly differentiated small cells with prominent dark nuclei and little cytoplasm, as well as several stem cell markers, such as erythropoietin (Epo), Epo receptor (EpoR), and CD133, are present in ocular VHL lesions. CXCR4, a CXC chemokine receptor is also expressed in retinal VHL hemangioblastomas. CONCLUSIONS These findings imply that VHL cells with LOH of the tumor suppressor gene, most likely originate from a hematopoietic/vascular lineage. Targeting these proteins and ischemic factors, not VEGF alone, may be a potential therapeutic approach for VHL-associated ocular hemangioblastomas.
Collapse
Affiliation(s)
- Chi-Chao Chan
- National Eye Institute, National Institutes of Health, Bethesda 20892-1857, USA.
| | | | | |
Collapse
|
444
|
Blouw B, Haase VH, Song H, Bergers G, Johnson RS. Loss of vascular endothelial growth factor expression reduces vascularization, but not growth, of tumors lacking the Von Hippel-Lindau tumor suppressor gene. Oncogene 2007; 26:4531-40. [PMID: 17297464 DOI: 10.1038/sj.onc.1210249] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Individuals bearing germ line mutations in the Von Hippel-Lindau (VHL) tumor suppressor gene are predisposed to the development of highly angiogenic tumors. This is correlated with an increased expression of the angiogenic factor vascular endothelial growth factor (VEGF) in these tumors, which is in part caused by elevated expression of the HIF-1 hypoxia inducible transcription factors. We created malignant astrocytes with genetic deletions of the VHL gene and implanted them in subcutaneous and intracranial sites; these sites are respectively vessel poor and vessel-rich tissues. When grown in a vessel poor site, VEGF expression in VHL null cells was important for both vascularization and tumor growth. However, when the same cells are grown in the vessel-rich intracranial environment, loss of VEGF expression reduces vascularization, but does not affect tumor growth. This indicates that antiangiogenic therapies for tumors that express high levels of angiogenic factors such as VEGF may vary in their efficacy, with potentially lowered effectiveness in sites, such as the brain, that are inherently vessel rich.
Collapse
Affiliation(s)
- B Blouw
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, La Jolla, California 92093-0377, USA
| | | | | | | | | |
Collapse
|
445
|
Affiliation(s)
- William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Howard Hughes Medical Institute, Chevy Chase, Maryland 20815;
| |
Collapse
|
446
|
Prindull G. Hemangioblasts representing a functional endothelio-hematopoietic entity in ontogeny, postnatal life, and CML neovasculogenesis. ACTA ACUST UNITED AC 2007; 1:277-84. [PMID: 17142866 DOI: 10.1385/scr:1:3:277] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The life-long interdependencies/interactions between hemato- and endotheliopoiesis suggest that they form a supplementary functional entity. This view is compatible with the concept of stem cell plasticity as a reversible continuum and is substantiated by the common hematopoietic-endothelial stem cell, i.e., hemangioblasts, with bidirectional, reversible gene transcription and persistence in postnatal life. Indeed, embryonal stem cells/hemangioblasts appear to form a reservior in the adult with the possibility of dedifferentiation of more differentiated progenitor cells back to hemangioblasts. The recent detection of BCR/ABL fusion proteins in endothelial cells during vascular neoangiogenesis in CML suggests that endothelial cells are part of the neoplastic clone, and extends the concept of a functional entity to include CML angiogenesis. Thus, hemangioblasts rather than committed hematopoietic stem cells appear to be target cells for the first oncogenic hit in CML, which could occur as early as during the first steps of embryonal stem cell differentiation towards hemato-endotheliopoiesis and/or in hemangioblasts persisting in adults. The relation of the other leukemias to hemangioblasts is not known.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Embryonic Stem Cells/metabolism
- Embryonic Stem Cells/pathology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Fusion Proteins, bcr-abl
- Gene Expression Regulation, Leukemic
- Hematopoiesis
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
Collapse
Affiliation(s)
- Gregor Prindull
- Pediatric Hematology/Oncology, University of Göttingen, Germany.
| |
Collapse
|
447
|
Abstract
Germ line VHL tumor suppressor gene loss-of-function mutations cause von Hippel-Lindau disease, which is associated with an increased risk of central nervous system hemangioblastomas, clear cell renal carcinomas, and pheochromocytomas. Somatic VHL mutations are also common in sporadic clear cell renal carcinomas. The VHL gene product, pVHL, is part of a ubiquitin ligase complex that targets the alpha-subunits of the heterodimeric transcription factor hypoxia-inducible factor (HIF) for polyubiquitylation, and hence, proteasomal degradation, when oxygen is available. pVHL-defective clear cell renal carcinomas overproduce a variety of mRNAs that are under the control of HIF, including the mRNAs that encode vascular endothelial growth factor, platelet-derived growth factor B, and transforming growth factor alpha. In preclinical models, down-regulation of HIF-alpha, especially HIF-2alpha, is both necessary and sufficient for renal tumor suppression by pVHL. These observations are probably relevant to the demonstrated clinical activity of vascular endothelial growth factor antagonists in clear cell renal carcinoma and form a foundation for the testing of additional agents that inhibit HIF, or HIF-responsive gene products, in this disease.
Collapse
Affiliation(s)
- William G Kaelin
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
448
|
Yan Q, Bartz S, Mao M, Li L, Kaelin WG. The hypoxia-inducible factor 2alpha N-terminal and C-terminal transactivation domains cooperate to promote renal tumorigenesis in vivo. Mol Cell Biol 2007; 27:2092-102. [PMID: 17220275 PMCID: PMC1820491 DOI: 10.1128/mcb.01514-06] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Hypoxia-inducible factor (HIF) is a heterodimeric transcription factor, consisting of an alpha subunit and a beta subunit, that controls cellular responses to hypoxia. HIFalpha contains two transcriptional activation domains called the N-terminal transactivation domain (NTAD) and the C-terminal transactivation domain (CTAD). HIFalpha is destabilized by prolyl hydroxylation catalyzed by EglN family members. In addition, CTAD function is inhibited by asparagine hydroxylation catalyzed by FIH1. Both hydroxylation reactions are linked to oxygen availability. The von Hippel-Lindau tumor suppressor protein (pVHL) is frequently mutated in kidney cancer and is part of the ubiquitin ligase complex that targets prolyl hydroxylated HIFalpha for destruction. Recent studies suggest that HIF2alpha plays an especially important role in promoting tumor formation by pVHL-defective renal carcinoma cells among the three HIFalpha paralogs. Here we dissected the relative contribution of the two HIF2alpha transactivation domains to hypoxic gene activation and renal carcinogenesis and investigated the regulation of the HIF2alpha CTAD by FIH1. We found that the HIF2alpha NTAD is capable of activating both artificial and naturally occurring HIF-responsive promoters in the absence of the CTAD. Moreover, we found that the HIF2alpha CTAD, in contrast to the HIF1alpha CTAD, is relatively resistant to the inhibitory effects of FIH1 under normoxic conditions and that, perhaps as a result, both the NTAD and CTAD cooperate to promote renal carcinogenesis in vivo.
Collapse
Affiliation(s)
- Qin Yan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
449
|
Hickey MM, Simon MC. Regulation of angiogenesis by hypoxia and hypoxia-inducible factors. Curr Top Dev Biol 2007; 76:217-57. [PMID: 17118268 DOI: 10.1016/s0070-2153(06)76007-0] [Citation(s) in RCA: 195] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Maintenance of oxygen homeostasis is critical for the survival of multicellular organs. As a result, both invertebrates and vertebrates have developed highly specialized mechanisms to sense changes in oxygen levels and to mount adequate cellular and systemic responses to these changes. Hypoxia, or low oxygen tension, occurs in physiological situations such as during embryonic development, as well as in pathological conditions such as ischemia, wound healing, and cancer. A primary effector of the adaptive response to hypoxia in mammals is the hypoxia-inducible factor (HIF) family of transcription regulators. These proteins activate the expression of a broad range of genes that mediate many of the responses to decreased oxygen concentration, including enhanced glucose uptake, increased red blood cell production, and the formation of new blood vessels via angiogenesis. This latter process is dynamic and results in the establishment of a mature vascular system that is indispensable for proper delivery of oxygen and nutrients to all cells in both normal tissue and hypoxic regions. Angiogenesis is essential for normal development and neoplastic disease as tumors must develop mechanisms to stimulate vascularization to meet increasing metabolic demands. The link between hypoxia and the regulation of angiogenesis is an area of intense research and the molecular details of this connection are still being elaborated. This chapter will provide an overview of current knowledge and highlight new insights into the importance of HIF and hypoxia in angiogenesis in both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Michele M Hickey
- Department of Cell and Molecular Biology, Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia 19104, USA
| | | |
Collapse
|
450
|
Evans AJ, Russell RC, Roche O, Burry TN, Fish JE, Chow VWK, Kim WY, Saravanan A, Maynard MA, Gervais ML, Sufan RI, Roberts AM, Wilson LA, Betten M, Vandewalle C, Berx G, Marsden PA, Irwin MS, Teh BT, Jewett MAS, Ohh M. VHL promotes E2 box-dependent E-cadherin transcription by HIF-mediated regulation of SIP1 and snail. Mol Cell Biol 2007; 27:157-69. [PMID: 17060462 PMCID: PMC1800649 DOI: 10.1128/mcb.00892-06] [Citation(s) in RCA: 210] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Revised: 06/27/2006] [Accepted: 09/27/2006] [Indexed: 12/13/2022] Open
Abstract
The product of the von Hippel-Lindau gene (VHL) acts as the substrate-recognition component of an E3 ubiquitin ligase complex that ubiquitylates the catalytic alpha subunit of hypoxia-inducible factor (HIF) for oxygen-dependent destruction. Although emerging evidence supports the notion that deregulated accumulation of HIF upon the loss of VHL is crucial for the development of clear-cell renal cell carcinoma (CC-RCC), the molecular events downstream of HIF governing renal oncogenesis remain unclear. Here, we show that the expression of a homophilic adhesion molecule, E-cadherin, a major constituent of epithelial cell junctions whose loss is associated with the progression of epithelial cancers, is significantly down-regulated in primary CC-RCC and CC-RCC cell lines devoid of VHL. Reintroduction of wild-type VHL in CC-RCC (VHL(-/-)) cells markedly reduced the expression of E2 box-dependent E-cadherin-specific transcriptional repressors Snail and SIP1 and concomitantly restored E-cadherin expression. RNA interference-mediated knockdown of HIFalpha in CC-RCC (VHL(-/-)) cells likewise increased E-cadherin expression, while functional hypoxia or expression of VHL mutants incapable of promoting HIFalpha degradation attenuated E-cadherin expression, correlating with the disengagement of RNA polymerase II from the endogenous E-cadherin promoter/gene. These findings reveal a critical HIF-dependent molecular pathway connecting VHL, an established "gatekeeper" of the renal epithelium, with a major epithelial tumor suppressor, E-cadherin.
Collapse
Affiliation(s)
- Andrew J Evans
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|