51
|
Zhang Q, Yang H. The Roles of VHL-Dependent Ubiquitination in Signaling and Cancer. Front Oncol 2012; 2:35. [PMID: 22649785 PMCID: PMC3355907 DOI: 10.3389/fonc.2012.00035] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/26/2012] [Indexed: 01/07/2023] Open
Abstract
The function of tumor suppressor VHL is compromised in the vast majority of clear cell renal cell carcinoma, and its mutations or loss of expression was causal for this disease. pVHL was found to be a substrate recognition subunit of an E3 ubiquitin ligase, and most of the tumor-derived mutations disrupt this function. pVHL was found to bind to the alpha subunits of hypoxia-inducible factor (HIF) and promote their ubiquitination and proteasomal degradation. Proline hydroxylation on key sites of HIFα provides the binding signal for pVHL E3 ligase complex. Beside HIFα, several other VHL targets have been identified, including activated epidermal growth factor receptor (EGFR), RNA polymerase II subunits RPB1 and hsRPB7, atypical protein kinase C (PKC), Sprouty2, β-adrenergic receptor II, and Myb-binding protein p160. HIFα is the most well studied substrate and has been proven to be critical for pVHL's tumor suppressor function, but the activated EGFR and PKC and other pVHL substrates might also be important for tumor growth and drug response. Their regulations by pVHL and their relevance to signaling and cancer are discussed.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, USA
| | | |
Collapse
|
52
|
Hillje AL, Worlitzer MMA, Palm T, Schwamborn JC. Neural stem cells maintain their stemness through protein kinase C ζ-mediated inhibition of TRIM32. Stem Cells 2012; 29:1437-47. [PMID: 21732497 DOI: 10.1002/stem.687] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Several studies over the last couple of years have delivered insights into the mechanisms that drive neuronal differentiation. However, the mechanisms that ensure the maintenance of stemness characteristics in neural stem cells over several rounds of cell divisions are still largely unknown. Here, we provide evidence that the neuronal fate determinant TRIM32 binds to the protein kinase C ζ. Through this interaction, TRIM32 is retained in the cytoplasm. However, during differentiation, this interaction is abrogated and TRIM32 translocates to the nucleus to initiate neuronal differentiation by targeting c-Myc for proteasomal degradation.
Collapse
Affiliation(s)
- Anna-Lena Hillje
- Westfälische Wilhelms-Universität Münster, ZMBE, Institute of Cell Biology, Stem Cell Biology and Regeneration Group, Münster, Germany
| | | | | | | |
Collapse
|
53
|
Okumura F, Matsuzaki M, Nakatsukasa K, Kamura T. The Role of Elongin BC-Containing Ubiquitin Ligases. Front Oncol 2012; 2:10. [PMID: 22649776 PMCID: PMC3355856 DOI: 10.3389/fonc.2012.00010] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 01/17/2012] [Indexed: 02/06/2023] Open
Abstract
The Elongin complex was originally identified as a positive regulator of RNA polymerase II and is composed of a transcriptionally active subunit (A) and two regulatory subunits (B and C). The Elongin BC complex enhances the transcriptional activity of Elongin A. “Classical” SOCS box-containing proteins interact with the Elongin BC complex and have ubiquitin ligase activity. They also interact with the scaffold protein Cullin (Cul) and the RING domain protein Rbx and thereby are members of the Cullin RING ligase (CRL) superfamily. The Elongin BC complex acts as an adaptor connecting Cul and SOCS box proteins. Recently, it was demonstrated that classical SOCS box proteins can be further divided into two groups, Cul2- and Cul5-type proteins. The classical SOCS box-containing protein pVHL is now classified as a Cul2-type protein. The Elongin BC complex containing CRL family is now considered two distinct protein assemblies, which play an important role in regulating a variety of cellular processes such as tumorigenesis, signal transduction, cell motility, and differentiation.
Collapse
Affiliation(s)
- Fumihiko Okumura
- Division of Biological Science, Graduate School of Science, Nagoya University Nagoya, Aichi, Japan
| | | | | | | |
Collapse
|
54
|
The changes of signal transduction pathways in hippocampal regions and postsynaptic densities after chronic cerebral hypoperfusion in rats. Brain Res 2012; 1429:9-17. [DOI: 10.1016/j.brainres.2011.10.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Revised: 10/11/2011] [Accepted: 10/12/2011] [Indexed: 11/15/2022]
|
55
|
Tootee A, Hasani- Ranjbar S. Von hippel-lindau disease: a new approach to an old problem. Int J Endocrinol Metab 2012; 10:619-24. [PMID: 23843833 PMCID: PMC3693632 DOI: 10.5812/ijem.4510] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 04/29/2012] [Accepted: 05/07/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Von Hippel-Lindau (VHL) disease is a hereditary, autosomal dominant syndrome which is manifested by a range of different benign and malignant tumors. This disease can present with different clinical presentations such as; retinal angioma (RA), hemangioblastoma (HB) of the central nervous system (CNS), pheochromocytoma (Pheo), and epididymal cystadenoma. Tumors are usually accompanied with cysts. OBJECTIVES As the disease can display different clinical presentations, which are mainly unspecific, and considering the importance of an early diagnosis and the proper and early management of it, this study was carried out to present a general overview of VHL. Moreover, the present article reviews screening methods and emphasizes the need for increasing the awareness of different health care professionals to diagnose and refer the patients in the early stages. MATERIALS AND METHODS A thorough search of internet medical databases, such as PubMed, was carried out on known or suggested; clinical presentations, pathogenesis, screening, causes and criteria for diagnosis of patients and their referrals. RESULTS Our research demonstrated that VHL is caused by a mutation in the von Hippel-Lindau (VHL) gene. It also showed that different screening methods can be utilized for the early diagnosis and referral of patients. Different clinical presentations of the disease are also elaborated in some detail and their treatment options are discussed. CONCLUSIONS Considering the need for a multidisciplinary approach to VHL, especially, given the number of cases which have been reported and diagnosed in Iran, it is of great importance that clinicians remain vigilant in order to identify cases that present with clinical characteristics of the disease, and that they are prompt in referring them to a multidisciplinary VHL clinic. It is also important to establish links with existing VHL Family Alliances and other related organizations around the world.
Collapse
Affiliation(s)
- Ali Tootee
- Tehran University of Medical Sciences, Endocrinology and Metabolism Research Institute, Shariati Hospital, Tehran, IR Iran
| | - Shirin Hasani- Ranjbar
- Tehran University of Medical Sciences, Endocrinology and Metabolism Research Institute, Shariati Hospital, Tehran, IR Iran
- Corresponding author: Shirin Hasani- Ranjbar, Endocrinology & Metabolism Research Institute, 5th Floor, Shariati Hospital, North Kargar Ave, 14114, Tehran, IR Iran. Tel.: +98-2188220037, Fax: +98-2188220052, E-mail:
| |
Collapse
|
56
|
|
57
|
Abstract
The cullin family of ubiquitin ligases can potentially assemble hundreds of RING-type E3 complexes (CRLs) by utilizing different substrate receptors that share common interaction domains. Cullin receptors dictate substrate specificity, and cullin-mediated substrate degradation controls a wide range of cellular processes, including proliferation, differentiation, and apoptosis. Dysregulation of cullin activity has been shown to contribute to oncogenesis through the accumulation of oncoproteins or the excessive degradation of tumor suppressors. In this review, we will discuss cullin complexes and their substrates, the regulatory pathways that affect cullin activity, and the mechanisms by which cullins may facilitate or inhibit carcinogenesis.
Collapse
Affiliation(s)
- Jennifer Lee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College and Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | | |
Collapse
|
58
|
Lim DL, Ko R, Pautler SE. Current understanding of the molecular mechanisms of kidney cancer: a primer for urologists. Can Urol Assoc J 2011; 1:S13-20. [PMID: 18542780 DOI: 10.5489/cuaj.63] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC), the fifth leading malignant condition for men and tenth for women, accounts for 3% of all malignancies in Canada. It is a heterogeneous epithelial malignancy with different subtypes and varied tumour biology. Although most cases of RCC are sporadic, up to 4% of patients have an inherited predisposition for the disease. In this article, we review the current molecular genetics of the different subtypes in hereditary and sporadic RCC. Significant developments in understanding the underlying genetic basis of RCC over the last 2 decades are attributed to intensive research about rare inherited renal cancer syndromes and the identification of the genes responsible for them. Many of these genes are also found in sporadic RCC. Understanding the molecular mechanisms involved in the pathogenesis of RCC has aided the development of molecular-targeted drugs for this disease.
Collapse
Affiliation(s)
- Darwin L Lim
- Divisions of Urology and Surgical Oncology, University of Western Ontario, London, Ont
| | | | | |
Collapse
|
59
|
Bonnet M, Flanagan JU, Chan DA, Lai EW, Nguyen P, Giaccia AJ, Hay MP. SAR studies of 4-pyridyl heterocyclic anilines that selectively induce autophagic cell death in von Hippel-Lindau-deficient renal cell carcinoma cells. Bioorg Med Chem 2011; 19:3347-56. [PMID: 21561782 PMCID: PMC3115526 DOI: 10.1016/j.bmc.2011.04.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/15/2011] [Accepted: 04/21/2011] [Indexed: 01/08/2023]
Abstract
We recently identified a class of pyridyl aniline thiazoles (PAT) that displayed selective cytotoxicity for von Hippel-Lindau (VHL) deficient renal cell carcinoma (RCC) cells in vitro and in vivo. Structure-activity relationship (SAR) studies were used to develop a comparative molecular field analysis (CoMFA) model that related VHL-selective potency to the three-dimensional arrangement of chemical features of the chemotype. We now report the further molecular alignment-guided exploration of the chemotype to discover potent and selective PAT analogues. The contribution of the central thiazole ring was explored using a series of five- and six-membered ring heterocyclic replacements to vary the electronic and steric interactions in the central unit. We also explored a positive steric CoMFA contour adjacent to the pyridyl ring using Pd-catalysed cross-coupling Suzuki-Miyaura, Sonogashira and nucleophilic displacement reactions to prepare of a series of aryl-, alkynyl-, alkoxy- and alkylamino-substituted pyridines, respectively. In vitro potency and selectivity were determined using paired RCC cell lines: the VHL-null cell line RCC4 and the VHL-positive cell line RCC4-VHL. Active analogues selectively induced autophagy in RCC4 cells. We have used the new SAR data to further develop the CoMFA model, and compared this to a 2D-QSAR method. Our progress towards realising the therapeutic potential of this chemotype as a targeted cytotoxic therapy for the treatment of RCC by exploiting the absence of the VHL tumour suppressor gene is reported.
Collapse
Affiliation(s)
- Muriel Bonnet
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Jack, U. Flanagan
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Denise A. Chan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Edwin W. Lai
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Phuong Nguyen
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P. Hay
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
60
|
Abstract
The correct establishment and maintenance of cell polarity are crucial for normal cell physiology and tissue homeostasis. Conversely, loss of cell polarity, tissue disorganisation and excessive cell growth are hallmarks of cancer. In this review, we focus on identifying the stages of tumoural development that are affected by the loss or deregulation of epithelial cell polarity. Asymmetric division has recently emerged as a major regulatory mechanism that controls stem cell numbers and differentiation. Links between cell polarity and asymmetric cell division in the context of cancer will be examined. Apical–basal polarity and cell–cell adhesion are tightly interconnected. Hence, how loss of cell polarity in epithelial cells may promote epithelial mesenchymal transition and metastasis will also be discussed. Altogether, we present the argument that loss of epithelial cell polarity may have an important role in both the initiation of tumourigenesis and in later stages of tumour development, favouring the progression of tumours from benign to malignancy.
Collapse
|
61
|
Epithelial cell polarity and tumorigenesis: new perspectives for cancer detection and treatment. Acta Pharmacol Sin 2011; 32:552-64. [PMID: 21499288 DOI: 10.1038/aps.2011.20] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Loss of cell-cell adhesion and cell polarity is commonly observed in tumors of epithelial origin and correlates with their invasion into adjacent tissues and formation of metastases. Growing evidence indicates that loss of cell polarity and cell-cell adhesion may also be important in early stage of cancer. In first part of this review, we delineate the current understanding of the mechanisms that establish and maintain the polarity of epithelial tissues and discuss the involvement of cell polarity and apical junctional complex components in tumor pathogenesis. In the second part we address the clinical significance of cell polarity and junctional complex components in cancer diagnosis and prognosis. Finally, we explore their potential use as therapeutic targets in the treatment of cancer.
Collapse
|
62
|
Li SH, Chun YS, Lim JH, Huang LE, Park JW. von Hippel-Lindau protein adjusts oxygen sensing of the FIH asparaginyl hydroxylase. Int J Biochem Cell Biol 2011; 43:795-804. [PMID: 21316481 DOI: 10.1016/j.biocel.2011.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 01/28/2011] [Accepted: 02/07/2011] [Indexed: 10/18/2022]
Abstract
Hypoxia inevitably develops in rapidly growing tumors and acts as an important microenvironment that forces changes in tumor behavior. Hypoxia-inducible factor 1α (HIF-1α) is activated during hypoxia and promotes the progression of malignancy by stimulating angiogenesis and by augmenting the ability of tumors to survive. In aerobic conditions, HIF-1α is destabilized by the PHD prolyl-hydroxylases that target HIF-1α for proteolysis via the von Hippel-Lindau protein (pVHL) and further inactivated by the FIH asparaginyl-hydroxylase that precludes the recruitment of transcription coactivators. Although HIF-1α degradation is well understood, little is known about how its transcriptional activity increases gradually in response to decreasing oxygen. In particular, it has been questioned how FIH having a high affinity for oxygen regulates the HIF-1α activity in moderate hypoxia. We here found that the HIF-1α-FIH interaction is disrupted in 1-5% oxygen. Both in vitro and in vivo binding analyses revealed that pVHL acts as an adaptor for FIH to bind HIF-1α. Furthermore, because the pVHL-FIH interaction depends on oxygen tension, the FIH-mediated inactivation of HIF-1α can be exquisitely regulated according to the severity of hypoxia. Based on these findings, we propose that pVHL fine-tunes the transcriptional activity of HIF-1α in graded oxygen tensions.
Collapse
Affiliation(s)
- Shan Hua Li
- Department of Pharmacology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | | | | | | | | |
Collapse
|
63
|
Regulation of the tumour suppressor Fbw7α by PKC-dependent phosphorylation and cancer-associated mutations. Biochem J 2010; 432:77-87. [DOI: 10.1042/bj20100799] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Fbw7 (F-box WD40 protein 7) is a major tumour suppressor, which mediates the degradation of several potent oncogenes. PKC (protein kinase C) comprises a serine/threonine kinase family that can promote transformation when dysregulated. In the present study, we investigated the relationship between Fbw7 and PKC. Multiple members of the PKC superfamily interact with the substrate-binding domain of Fbw7. However, we find no evidence for Fbw7-mediated degradation of PKC. Instead, we demonstrate that Fbw7 is a novel substrate for PKC. Two residues within the isoform-specific N-terminus of Fbw7α are phosphorylated in a PKC-dependent manner, both in vitro and in mammalian cells (Ser10 and Ser18). Mutational analyses reveal that phosphorylation of Fbw7α at Ser10 can regulate its nuclear localization. Cancer-associated mutations in nearby residues (K11R and the addition of a proline residue at position 16) influence Fbw7α localization in a comparable manner, suggesting that mislocalization of this protein may be of pathological significance. Together these results provide evidence for both physical and functional interactions between the PKC and Fbw7 families, and yield insights into the isoform-specific regulation of Fbw7α.
Collapse
|
64
|
Abstract
β-cells sense glucose and secrete appropriate amounts of insulin by coupling glucose uptake and glycolysis with quantitative ATP production via mitochondrial oxidative pathways. Therefore, oxidative phosphorylation is essential for normal β-cell function. Multiple cell types adapt to hypoxia by inducing a transcriptional programme coordinated by the transcription factor hypoxia-inducible factor (HIF). HIF activity is regulated by the von Hippel-Lindau (Vhl) protein, which targets the HIFα subunit for proteasomal degradation in the presence of oxygen. Several recent studies have shown that Vhl deletion in β-cells results in Hif1α activation, impaired glucose-stimulated insulin secretion (GSIS) and glucose intolerance. This was found to be because of alterations in β-cell gene expression inducing a switch from aerobic glucose metabolism to anaerobic glycolysis, thus disrupting the GSIS triggering pathway. Situations in which islets may become hypoxic are discussed, in particular islet transplantation which has been reported to cause islet hypoxia because of an inadequate blood supply post-transplant. Aside from this principal role for HIF in negatively regulating β-cell glucose sensing, other aspects of hypoxia signalling are discussed including β-cell differentiation, development and vascularization. In conclusion, recent studies clearly show that hypoxia response mechanisms can negatively impact on glucose sensing mechanisms in the β-cell and this has the potential to impair β-cell function in a number of physiological and clinical situations.
Collapse
Affiliation(s)
- J Cantley
- Faculty of Medicine, Rayne Institute, University College London, London, UK.
| | | | | | | |
Collapse
|
65
|
Gao C, Cao W, Bao L, Zuo W, Xie G, Cai T, Fu W, Zhang J, Wu W, Zhang X, Chen YG. Autophagy negatively regulates Wnt signalling by promoting Dishevelled degradation. Nat Cell Biol 2010; 12:781-90. [DOI: 10.1038/ncb2082] [Citation(s) in RCA: 300] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 07/05/2010] [Indexed: 12/18/2022]
|
66
|
Abstract
Multiple epidemiologic studies have linked the development of renal cancer to obesity. In this chapter, we begin with a review of selected population studies, followed by recent mechanistic discoveries that further link lipid deregulation to the RCC development. The upregulation of leptin and downregulation of adiponectin pathways in obesity fit well with our molecular understanding of RCC pathogenesis. In addition, two forms of hereditary RCC involve proteins, Folliculin and TRC8, that are positioned to coordinately regulate lipid and protein biosynthesis. Both of these biosynthetic pathways have important downstream consequences on HIF-1/2alpha levels and angiogenesis, key aspects in the disease pathogenesis. The role of lipid biology and its interface with protein translation regulation represents a new dimension in RCC research with potential therapeutic implications.
Collapse
Affiliation(s)
- Harry A Drabkin
- Department of Medicine and Division of Hematology-Oncology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
67
|
Hay MP, Turcotte S, Flanagan JU, Bonnet M, Chan DA, Sutphin PD, Nguyen P, Giaccia AJ, Denny WA. 4-Pyridylanilinothiazoles that selectively target von Hippel-Lindau deficient renal cell carcinoma cells by inducing autophagic cell death. J Med Chem 2010; 53:787-97. [PMID: 19994864 DOI: 10.1021/jm901457w] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Renal cell carcinomas (RCC) are refractory to standard therapy with advanced RCC having a poor prognosis; consequently treatment of advanced RCC represents an unmet clinical need. The von Hippel-Lindau (VHL) tumor suppressor gene is mutated or inactivated in a majority of RCCs. We recently identified a 4-pyridyl-2-anilinothiazole (PAT) with selective cytotoxicity against VHL-deficient renal cells mediated by induction of autophagy and increased acidification of autolysosomes. We report exploration of structure-activity relationships (SAR) around this PAT lead. Analogues with substituents on each of the three rings, and various linkers between rings, were synthesized and tested in vitro using paired RCC4 cell lines. A contour map describing the relative spatial contributions of different chemical features to potency illustrates a region, adjacent to the pyridyl ring, with potential for further development. Examples probing this domain validated this approach and may provide the opportunity to develop this novel chemotype as a targeted approach to the treatment of RCC.
Collapse
Affiliation(s)
- Michael P Hay
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Haase VH. The VHL tumor suppressor: master regulator of HIF. Curr Pharm Des 2010; 15:3895-903. [PMID: 19671042 DOI: 10.2174/138161209789649394] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 07/01/2009] [Indexed: 12/11/2022]
Abstract
Hypoxia-Inducible Factors (HIFs) are heterodimeric oxygen-sensitive basic helix-loop-helix transcription factors that play central roles in cellular adaptation to low oxygen environments. The von-Hippel Lindau tumor suppressor (pVHL) is the substrate recognition component of an E3 ubiquitin ligase and functions as a master regulator of HIF activity by targeting the hydroxylated HIF-alpha subunit for ubiquitylation and rapid proteasomal degradation under normoxic conditions. Mutations in pVHL can be found in familial and sporadic hemangioblastomas, clear cell carcinomas of the kidney, pheochromocytomas and inherited forms of erythrocytosis, illustrating the importance of disrupted molecular oxygen sensing in the pathogenesis of these diseases. Tissue-specific gene targeting of pVHL in mice has demonstrated that efficient execution of HIF proteolysis is critically important for normal tissue physiology, and has provided novel insights into the functional consequences of HIF activation on the cellular and tissue level. Here we focus on the contribution of individual HIF transcription factors to the development of VHL phenotypes and discuss how the pVHL/HIF axis could be exploited pharmacologically.
Collapse
Affiliation(s)
- Volker H Haase
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
69
|
Tao T, Ji Y, Cheng C, Yang H, Liu H, Sun L, Qin Y, Yang J, Wang H, Shen A. Tumor necrosis factor-alpha inhibits Schwann cell proliferation by up-regulating Src-suppressed protein kinase C substrate expression. J Neurochem 2009; 111:647-55. [DOI: 10.1111/j.1471-4159.2009.06346.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
70
|
Abstract
Protein kinases are important regulators of intracellular signal transduction pathways and play critical roles in diverse cellular functions. Once a protein kinase is activated, its activity is subsequently downregulated through a variety of mechanisms. Accumulating evidence indicates that the activation of protein kinases commonly initiates their downregulation via the ubiquitin/proteasome pathway. Failure to regulate protein kinase activity or expression levels can cause human diseases.
Collapse
Affiliation(s)
- Zhimin Lu
- Department of Neuro-Oncology and Molecular and Cellular Oncology, University of Texas M. D. Anderson Cancer, Houston, TX 77030, USA.
| | | |
Collapse
|
71
|
Guo Y, Schoell MC, Freeman RS. The von Hippel-Lindau protein sensitizes renal carcinoma cells to apoptotic stimuli through stabilization of BIM(EL). Oncogene 2009; 28:1864-74. [PMID: 19305426 PMCID: PMC2701369 DOI: 10.1038/onc.2009.35] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
von Hippel-Lindau (VHL) disease is caused by germ-line mutations in the VHL tumor suppressor gene and is the most common cause of inherited renal cell carcinoma (RCC). Mutations in the VHL gene also occur in a large majority of sporadic cases of clear-cell RCC, which have high intrinsic resistance to chemotherapy and radiotherapy. Here we show that VHL-deficient RCC cells express lower levels of the proapoptotic Bcl-2 family protein BIM(EL) and are more resistant to etoposide and UV radiation-induced death compared to the same cells stably expressing the wild-type VHL protein (pVHL). Reintroducing pVHL into VHL-null cells increased the half-life of BIM(EL) protein without affecting its mRNA expression, and overexpressing pVHL inhibited BIM(EL) polyubiquitination. Suppressing pVHL expression with RNA interference resulted in a decrease in BIM(EL) protein and a corresponding decrease in the sensitivity of RCC cells to apoptotic stimuli. Directly inhibiting BIM(EL) expression in pVHL-expressing RCC cells caused a similar decrease in cell death. These results demonstrate that pVHL acts to promote BIM(EL) protein stability in RCC cells, and that destabilization of BIM(EL) in the absence of pVHL contributes to the increased resistance of VHL-null RCC cells to certain apoptotic stimuli.
Collapse
Affiliation(s)
- Y Guo
- University of Rochester School of Medicine, NY 14642, USA
| | | | | |
Collapse
|
72
|
Abstract
Cancer is the result of the deregulation of cell proliferation and cell migration. In advanced tumors, cells invade the surrounding tissue and eventually form metastases. This is particularly evident in carcinomas in which epithelial cells have undergone epithelial-mesenchymal transition. Increased cell migration often correlates with a weakening of intercellular interactions. Junctions between neighboring epithelial cells are required to establish and maintain baso-apical polarity, suggesting that not only loss of cell-cell adhesion but also alteration of cell polarity is involved during invasion. Accordingly, perturbation of cell polarity is an important hallmark of advanced invasive tumors. Cell polarity is also essential for cell migration. Indeed, a front-rear polarity axis has first to be generated to allow a cell to migrate. Because cells migrate during invasion, cell polarity is not completely lost. Instead, polarity is modified. From a nonmigrating baso-apically polarized epithelial phenotype, cells acquire a polarized migrating mesenchymal phenotype. The aim of this review is to highlight the molecular relationship between the control of cell polarity and the regulation of cell motility during oncogenesis.
Collapse
Affiliation(s)
- S Etienne-Manneville
- Cell polarity and migration group, Institut Pasteur and CNRS URA 2582, Paris cedex 15, France.
| |
Collapse
|
73
|
Abstract
In the past 20 years, the discovery and characterization of the molecular machinery that controls cellular polarization have enabled us to achieve a better understanding of many biological processes. Spatial asymmetry or establishment of cell polarity during embryogenesis, epithelial morphogenesis, neuronal differentiation, and migration of fibroblasts and T cells are thought to rely on a small number of evolutionarily conserved proteins and pathways. Correct polarization is crucial for normal cell physiology and tissue homeostasis, and is lost in cancer. Thus, cell polarity signaling is likely to have an important function in tumor progression. Recent findings have identified a regulator of cell polarity, the Par complex, as an important signaling node in tumorigenesis. In normal cell types, the Par complex is part of the molecular machinery that regulates cell polarity and maintains normal cell homeostasis. As such, the polarity regulators are proposed to have a tumor suppressor function, consistent with the loss of polarity genes associated with hyperproliferation in Drosophila melanogaster. However, recent studies showing that some members of this complex also display pro-oncogenic activities suggest a more complex regulation of the polarity machinery during cellular transformation. Here, we examine the existing data about the different functions of the Par complex. We discuss how spatial restriction, binding partners and substrate specificity determine the signaling properties of Par complex proteins. A better understanding of these processes will very likely shed some light on how the Par complex can switch from a normal polarity regulation function to promotion of transformation downstream of oncogenes.
Collapse
|
74
|
Harten SK, Esteban MA, Maxwell PH. Identification of novel VHL regulated genes by transcriptomic analysis of RCC10 renal carcinoma cells. ACTA ACUST UNITED AC 2009; 49:43-52. [DOI: 10.1016/j.advenzreg.2008.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
75
|
Harten SK, Shukla D, Barod R, Hergovich A, Balda MS, Matter K, Esteban MA, Maxwell PH. Regulation of renal epithelial tight junctions by the von Hippel-Lindau tumor suppressor gene involves occludin and claudin 1 and is independent of E-cadherin. Mol Biol Cell 2008; 20:1089-101. [PMID: 19073886 DOI: 10.1091/mbc.e08-06-0566] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Epithelial-to-mesenchymal transitions (EMT) are important in renal development, fibrosis, and cancer. Loss of function of the tumor suppressor VHL leads to many features of EMT, and it has been hypothesized that the pivotal mediator is down-regulation of the adherens junction (AJ) protein E-cadherin. Here we show that VHL loss-of-function also has striking effects on the expression of the tight junction (TJ) components occludin and claudin 1 in vitro in VHL-defective clear cell renal cell carcinoma (CCRCC) cells and in vivo in VHL-defective sporadic CCRCCs (compared with normal kidney). Occludin is also down-regulated in premalignant foci in kidneys from patients with germline VHL mutations, consistent with a contribution to CCRCC initiation. Reexpression of E-cadherin was sufficient to restore AJ but not TJ assembly, indicating that the TJ defect is independent of E-cadherin down-regulation. Additional experiments show that activation of hypoxia inducible factor (HIF) contributes to both TJ and AJ abnormalities, thus the VHL/HIF pathway contributes to multiple aspects of the EMT phenotype that are not interdependent. Despite the independent nature of the defects, we show that treatment with the histone deacetylase inhibitor sodium butyrate, which suppresses HIF activation, provides a method for reversing EMT in the context of VHL inactivation.
Collapse
Affiliation(s)
- Sarah K Harten
- Division of Medicine, Rayne Institute, University College London, WC1E 6JJ, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Cantley J, Selman C, Shukla D, Abramov AY, Forstreuter F, Esteban MA, Claret M, Lingard SJ, Clements M, Harten SK, Asare-Anane H, Batterham RL, Herrera PL, Persaud SJ, Duchen MR, Maxwell PH, Withers DJ. Deletion of the von Hippel-Lindau gene in pancreatic beta cells impairs glucose homeostasis in mice. J Clin Invest 2008; 119:125-35. [PMID: 19065050 PMCID: PMC2613475 DOI: 10.1172/jci26934] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Accepted: 10/29/2008] [Indexed: 01/06/2023] Open
Abstract
Defective insulin secretion in response to glucose is an important component of the beta cell dysfunction seen in type 2 diabetes. As mitochondrial oxidative phosphorylation plays a key role in glucose-stimulated insulin secretion (GSIS), oxygen-sensing pathways may modulate insulin release. The von Hippel-Lindau (VHL) protein controls the degradation of hypoxia-inducible factor (HIF) to coordinate cellular and organismal responses to altered oxygenation. To determine the role of this pathway in controlling glucose-stimulated insulin release from pancreatic beta cells, we generated mice lacking Vhl in pancreatic beta cells (betaVhlKO mice) and mice lacking Vhl in the pancreas (PVhlKO mice). Both mouse strains developed glucose intolerance with impaired insulin secretion. Furthermore, deletion of Vhl in beta cells or the pancreas altered expression of genes involved in beta cell function, including those involved in glucose transport and glycolysis, and isolated betaVhlKO and PVhlKO islets displayed impaired glucose uptake and defective glucose metabolism. The abnormal glucose homeostasis was dependent on upregulation of Hif-1alpha expression, and deletion of Hif1a in Vhl-deficient beta cells restored GSIS. Consistent with this, expression of activated Hif-1alpha in a mouse beta cell line impaired GSIS. These data suggest that VHL/HIF oxygen-sensing mechanisms play a critical role in glucose homeostasis and that activation of this pathway in response to decreased islet oxygenation may contribute to beta cell dysfunction.
Collapse
Affiliation(s)
- James Cantley
- Centre for Diabetes and Endocrinology, Faculty of Medicine, Rayne Institute, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
The epithelial polarity program: machineries involved and their hijacking by cancer. Oncogene 2008; 27:6939-57. [DOI: 10.1038/onc.2008.345] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
78
|
Abstract
Once engaged by soluble or matrix-anchored ligands, cell surface proteins are commonly sorted to lysosomal degradation through several endocytic pathways. Defective vesicular trafficking of growth factor receptors, as well as unbalanced recycling of integrin- and cadherin-based adhesion complexes, has emerged in the past 5 years as a multifaceted hallmark of malignant cells. In line with the cooperative nature of endocytic machineries, multiple oncogenic alterations underlie defective endocytosis, such as altered ubiquitylation (Cbl and Nedd4 ubiquitin ligases, for example), altered cytoskeletal interactions and alterations to Rab family members. Pharmaceutical interception of the propensity of tumour cells to derail their signalling and their adhesion receptors may constitute a novel target for cancer therapy.
Collapse
Affiliation(s)
- Yaron Mosesson
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | |
Collapse
|
79
|
Abstract
The von Hippel-Lindau disease is caused by inactivating germline mutations of the VHL tumour suppressor gene and is associated with an increased risk of a variety of tumours in an allele-specific manner. The role of the heterodimeric transcription factor hypoxia-inducible factor (HIF) in the pathogenesis of VHL-defective tumours has been more firmly established during the past 5 years. In addition, there is now a greater appreciation of HIF-independent VHL functions that are relevant to tumour development, including maintenance of the primary cilium, regulation of extracellular matrix formation and turnover, and modulation of cell death in certain cell types following growth factor withdrawal or in response to other forms of stress.
Collapse
Affiliation(s)
- William G Kaelin
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815-6789, USA.
| |
Collapse
|
80
|
Santos N, Reiter JF. Building it up and taking it down: the regulation of vertebrate ciliogenesis. Dev Dyn 2008; 237:1972-81. [PMID: 18435467 DOI: 10.1002/dvdy.21540] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Primary cilia project from the surface of most vertebrate cells, and function in sensation and signaling during both development and adult tissue homeostasis. Mounting evidence links ciliary defects with a wide variety of diseases, underscoring the importance of understanding how these dynamic organelles are assembled and maintained. However, despite their physiological and clinical relevance, the logic and machinery that regulate ciliogenesis remain largely enigmatic. Here, we summarize emerging data that connect the assembly and disassembly of the primary cilium to cell cycle progression and we examine how determinants of cell architecture, including the planar cell polarity pathway, may regulate ciliogenesis. Additionally, identification of the genes underlying diverse ciliopathies in human patients is shedding light on the regulation of the formation of this complex organelle.
Collapse
Affiliation(s)
- Nicole Santos
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94158-2324, USA
| | | |
Collapse
|
81
|
Basu A, Contreras AG, Datta D, Flynn E, Zeng L, Cohen HT, Briscoe DM, Pal S. Overexpression of vascular endothelial growth factor and the development of post-transplantation cancer. Cancer Res 2008; 68:5689-98. [PMID: 18632621 DOI: 10.1158/0008-5472.can-07-6603] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer is an increasing and major problem after solid organ transplantation. In part, the increased cancer risk is associated with the use of immunosuppressive agents, especially calcineurin inhibitors. We propose that the effect of calcineurin inhibitors on the expression of vascular endothelial growth factor (VEGF) leads to an angiogenic milieu that favors tumor growth. Here, we used 786-0 human renal cancer cells to investigate the effect of cyclosporine (CsA) on VEGF expression. Using a full-length VEGF promoter-luciferase construct, we found that CsA markedly induced VEGF transcriptional activation through the protein kinase C (PKC) signaling pathway, specifically involving PKC zeta and PKC delta isoforms. Moreover, CsA promoted the association of PKC zeta and PKC delta with the transcription factor Sp1 as observed by immunoprecipitation assays. Using promoter deletion constructs, we found that CsA-mediated VEGF transcription was primarily Sp1 dependent. Furthermore, CsA-induced and PKC-Sp1-mediated VEGF transcriptional activation was partially inhibited by von Hippel-Lindau protein. CsA also promoted the progression of human renal tumors in vivo, wherein VEGF is overexpressed. Finally, to evaluate the in vivo significance of CsA-induced VEGF overexpression in terms of post-transplantation tumor development, we injected CT26 murine carcinoma cells (known to form angiogenic tumors) into mice with fully MHC mismatched cardiac transplants. We observed that therapeutic doses of CsA increased tumor size and VEGF mRNA expression and also enhanced tumor angiogenesis. However, coadministration of a blocking anti-VEGF antibody inhibited this CsA-mediated tumor growth. Collectively, these findings define PKC-mediated VEGF transcriptional activation as a key component in the progression of CsA-induced post-transplantation cancer.
Collapse
Affiliation(s)
- Aninda Basu
- Division of Nephrology, Children's Hospital Boston, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Lee M, Vasioukhin V. Cell polarity and cancer--cell and tissue polarity as a non-canonical tumor suppressor. J Cell Sci 2008; 121:1141-50. [PMID: 18388309 DOI: 10.1242/jcs.016634] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Correct establishment and maintenance of cell polarity is required for the development and homeostasis of all metazoans. Cell-polarity mechanisms are responsible not only for the diversification of cell shapes but also for regulation of the asymmetric cell divisions of stem cells that are crucial for their correct self-renewal and differentiation. Disruption of cell polarity is a hallmark of cancer. Furthermore, recent evidence indicates that loss of cell polarity is intimately involved in cancer: several crucial cell-polarity proteins are known proto-oncogenes or tumor suppressors, basic mechanisms of cell polarity are often targeted by oncogenic signaling pathways, and deregulation of asymmetric cell divisions of stem or progenitor cells may be responsible for abnormal self-renewal and differentiation of cancer stem cells. Data from in vivo and three-dimensional (3D) cell-culture models demonstrate that tissue organization attenuates the phenotypic outcome of oncogenic signaling. We suggest that polarized 3D tissue organization uses cell-cell and cell-substratum adhesion structures to reinforce and maintain the cell polarity of pre-cancerous cells. In this model, polarized 3D tissue organization functions as a non-canonical tumor suppressor that prevents the manifestation of neoplastic features in mutant cells and, ultimately, suppresses tumor development and progression.
Collapse
Affiliation(s)
- Minhui Lee
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N., C3-168, Seattle, WA 98109, USA
| | | |
Collapse
|
83
|
Abstract
Protein kinase C (PKC) is a family of kinases that plays diverse roles in many cellular functions, notably proliferation, differentiation, and cell survival. PKC is processed by phosphorylation and regulated by cofactor binding and subcellular localization. Extensive detail is available on the molecular mechanisms that regulate the maturation, activation, and signaling of PKC. However, less information is available on how signaling is terminated both from a global perspective and isozyme-specific differences. To target PKC therapeutically, various ATP-competitive inhibitors have been developed, but this method has problems with specificity. One possible new approach to developing novel, specific therapeutics for PKC would be to target the signaling termination pathways of the enzyme. This review focuses on the new developments in understanding how PKC signaling is terminated and how current drug therapies as well as information obtained from the recent elucidation of various PKC structures and down-regulation pathways could be used to develop novel and specific therapeutics for PKC.
Collapse
Affiliation(s)
- Christine M. Gould
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093-0721
| | - Alexandra C. Newton
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093-0721
| |
Collapse
|
84
|
Turcotte S, Chan DA, Sutphin PD, Hay MP, Denny WA, Giaccia AJ. A molecule targeting VHL-deficient renal cell carcinoma that induces autophagy. Cancer Cell 2008; 14:90-102. [PMID: 18598947 PMCID: PMC2819422 DOI: 10.1016/j.ccr.2008.06.004] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 02/02/2008] [Accepted: 06/09/2008] [Indexed: 02/07/2023]
Abstract
Renal cell carcinomas (RCCs) are refractory to standard therapies. The von Hippel-Lindau (VHL) tumor suppressor gene is inactivated in 75% of RCCs. By screening for small molecules selectively targeting VHL-deficient RCC cells, we identified STF-62247. STF-62247 induces cytotoxicity and reduces tumor growth of VHL-deficient RCC cells compared to genetically matched cells with wild-type VHL. STF-62247-stimulated toxicity occurs in a HIF-independent manner through autophagy. Reduction of protein levels of essential autophagy pathway components reduces sensitivity of VHL-deficient cells to STF-62247. Using a yeast deletion pool, we show that loss of proteins involved in Golgi trafficking increases killing by STF-62247. Thus, we have found a small molecule that selectively induces cell death in VHL-deficient cells, representing a paradigm shift for targeted therapy.
Collapse
Affiliation(s)
- Sandra Turcotte
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Denise A Chan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Patrick D Sutphin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Michael P Hay
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand, Private Bag 92019, Auckland, New Zealand
| | - William A Denny
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand, Private Bag 92019, Auckland, New Zealand
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
- Corresponding author. Amato J Giaccia,
| |
Collapse
|
85
|
Abstract
Mutations in the von Hippel-Lindau tumor suppressor gene VHL occur in various inherited and sporadically occurring tumors. The protein encoded by VHL--pVHL--bears no known enzymatic activities but interacts with numerous protein partners. With the identification of distinct pVHL-containing multiprotein complexes, a refined portrait of pVHL tumor suppressor function has arisen. In general, pVHL acts as a multipurpose adaptor protein that controls a diverse array of gene expression programs, as well as extracellular matrix assembly and microtubule-based processes, by linking various target proteins to appropriate enzymatic activities. These findings provide an evermore complex but coherent view of how pVHL functions molecularly and of the consequences of dysregulation of these diverse molecular activities on tumor formation.
Collapse
Affiliation(s)
- Ian J Frew
- Institute of Cell Biology, Eidgenössische Technische Hochschule Zurich, 8093 Zurich, Switzerland
| | | |
Collapse
|
86
|
Symmetrically dividing cell specific division axes alteration observed in proteasome depleted C. elegans embryo. Mech Dev 2008; 125:743-55. [PMID: 18502617 DOI: 10.1016/j.mod.2008.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Revised: 04/08/2008] [Accepted: 04/09/2008] [Indexed: 11/21/2022]
Abstract
A fertilised Caenorhabditis elegans embryo shows an invariable pattern of cell division and forms a multicellular body where each cell locates to a defined position. Mitotic spindle orientation is determined by several preceding events including the migration of duplicated centrosomes on a nucleus and the rotation of nuclear-centrosome complex. Cell polarity is the dominant force driving nuclear-centrosome rotation and setting the mitotic spindle axis in parallel with the polarity axis during asymmetric cell division. It is reasonable that there is no nuclear-centrosome rotation in symmetrically dividing blastomeres, but the mechanism(s) which suppress rotation in these cells have been proposed because the rotations occur in some polarity defect embryos. Here we show the nuclear-centrosome rotation can be induced by depletion of RPN-2, a regulatory subunit of the proteasome. In these embryos, cell polarity is established normally and both asymmetrically and symmetrically dividing cells are generated through asymmetric cell divisions. The nuclear-centrosome rotations occurred normally in the asymmetrically dividing cell lineage, but also induced in symmetrically dividing daughter cells. Interestingly, we identified RPN-2 as a binding protein of PKC-3, one of critical elements for establishing cell polarity during early asymmetric cell divisions. In addition to asymmetrically dividing cells, PKC-3 is also expressed in symmetrically dividing cells and a role to suppress nuclear-centrosome rotation has been anticipated. Our data suggest that the expression of RPN-2 is involved in the mechanism to suppress nuclear-centrosome rotation in symmetrically dividing cells and it may work in cooperation with PKC-3.
Collapse
|
87
|
Costa LJ, Drabkin HA. Renal cell carcinoma: new developments in molecular biology and potential for targeted therapies. Oncologist 2008; 12:1404-15. [PMID: 18165617 DOI: 10.1634/theoncologist.12-12-1404] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Renal cell carcinoma (RCC) affects 38,000 individuals in the U.S. yearly. Seventy-five percent of cases are clear-cell carcinomas, and a majority is driven by dysfunction of the von Hippel-Lindau (VHL) gene. VHL loss of function and other non-VHL pathways leading to RCC share aberrant activation of the hypoxic response, such as upregulation of vascular endothelial growth factor (VEGF) and consequent neoangiogenesis. Metastatic RCC has been notoriously resistant to therapy. For decades, its treatment has been based on nephrectomy and limited use of toxic and often inefficient immunotherapy with interleukin-2 or interferon-alpha. However, new biologic agents are beginning to break the resistance barrier. Small-molecule multikinase inhibitors that target VEGF receptors (sunitinib and sorafenib) have a favorable toxicity profile and can prolong time to progression and preserve quality of life when used in newly diagnosed or previously treated patients. The anti-VEGF antibody bevacizumab enhances the response rate and prolongs disease control when added to interferon-alpha. Temsirolimus, a mammalian target of rapamycin inhibitor, prolongs the survival duration of patients with poor-risk disease. Despite three new drugs being approved for RCC in the past 2 years, responses are mostly partial and of limited duration. Multiple new drugs and drug combinations are undergoing clinical trials and will likely impact the treatment of RCC in future years. Compounds found to be active in the metastatic setting are now being tried in earlier stage disease in an attempt to improve curability. However, no method has yet been validated to predict patient response to these newer treatments.
Collapse
Affiliation(s)
- Luciano J Costa
- University of Colorado Health Sciences Center, Denver, CO, USA
| | | |
Collapse
|
88
|
The von Hippel-Lindau tumor suppressor protein and Egl-9-Type proline hydroxylases regulate the large subunit of RNA polymerase II in response to oxidative stress. Mol Cell Biol 2008; 28:2701-17. [PMID: 18285459 DOI: 10.1128/mcb.01231-07] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human renal clear cell carcinoma (RCC) is frequently associated with loss of the von Hippel-Lindau (VHL) tumor suppressor (pVHL), which inhibits ubiquitylation and degradation of the alpha subunits of hypoxia-inducible transcription factor. pVHL also ubiquitylates the large subunit of RNA polymerase II, Rpb1, phosphorylated on serine 5 (Ser5) within the C-terminal domain (CTD). A hydroxylated proline 1465 within an LXXLAP motif located N-terminal to the CTD allows the interaction of Rpb1 with pVHL. Here we report that in RCC cells, pVHL regulates expression of Rpb1 and is necessary for low-grade oxidative-stress-induced recruitment of Rpb1 to the DNA-engaged fraction and for its P1465 hydroxylation, phosphorylation, and nondegradative ubiquitylation. Egln-9-type prolyl hydroxylases, PHD1 and PHD2, coimmunoprecipitated with Rpb1 in the chromatin fraction of VHL(+) RCC cells in response to oxidative stress, and PHD1 was necessary for P1465 hydroxylation while PHD2 had an inhibitory effect. P1465 hydroxylation was required for oxidative-stress-induced Ser5 phosphorylation of Rpb1. Importantly, overexpression of wild-type Rpb1 stimulated formation of kidney tumors by VHL(+) cells, and this effect was abolished by P1465A mutation of Rpb1. These data indicate that through this novel pathway involving P1465 hydroxylation and Ser5 phosphorylation of Rbp1, pVHL may regulate tumor growth.
Collapse
|
89
|
Kapitsinou PP, Haase VH. The VHL tumor suppressor and HIF: insights from genetic studies in mice. Cell Death Differ 2008; 15:650-9. [PMID: 18219317 DOI: 10.1038/sj.cdd.4402313] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The von Hippel-Lindau tumor suppressor gene product, pVHL, functions as the substrate recognition component of an E3-ubiquitin ligase, which targets the oxygen-sensitive alpha-subunit of hypoxia-inducible factor (HIF) for rapid proteasomal degradation under normoxic conditions and as such plays a central role in molecular oxygen sensing. Mutations in pVHL can be found in familial and sporadic clear cell carcinomas of the kidney, hemangioblastomas of the retina and central nervous system, and pheochromocytomas, underscoring its gatekeeper function in the pathogenesis of these tumors. Tissue-specific gene targeting of VHL in mice has demonstrated that efficient execution of pVHL-mediated HIF proteolysis under normoxia is fundamentally important for survival, proliferation, differentiation and normal physiology of many cell types, and has provided novel insights into the biological function of individual HIF transcription factors. In this review, we discuss the role of HIF in the development of the VHL phenotype.
Collapse
Affiliation(s)
- P P Kapitsinou
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
90
|
Rathmell WK, Chen S. VHL inactivation in renal cell carcinoma: implications for diagnosis, prognosis and treatment. Expert Rev Anticancer Ther 2008; 8:63-73. [PMID: 18095884 DOI: 10.1586/14737140.8.1.63] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) provides a tumor paradigm for the integration of genetics, molecular biology, therapeutic target validation and the introduction of high-impact treatment strategies. Most cases of sporadic as well as familial ccRCC acquire somatic inactivating mutations of the von Hippel-Lindau tumor-suppressor gene, VHL. pVHL, VHL gene product and a protein member of the E3 ubiquitin ligase family, acts in normal cells to direct the degradation and clearance of the hypoxia inducible factor (HIF)alpha transcription factor family, such that in its absence, as in ccRCC, the HIF proteins stabilize, accumulate to supraphysiologic levels and activate the transcription of genes such as VEGF and PDGF, which contributes substantially to the physiology of the tumor, and has been assessed indirectly as a prognostic factor. Molecularly targeted therapy blocking components of this pathway has been successfully introduced to the clinic with a substantive impact on clinical parameters of RCC. This review will examine the regulation of these molecular pathways in RCC and discuss the impact on the clinical management of patients with RCC.
Collapse
Affiliation(s)
- W Kimryn Rathmell
- The University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, 450 West Drive, Campus Box 7295, Chapel Hill, NC 27599-7295, USA.
| | | |
Collapse
|
91
|
Abstract
The development of cancer is a multistep process in which the DNA of a single cell accumulates mutations in genes that control essential cellular processes. Loss of cell-cell adhesion and cell polarity is commonly observed in advanced tumours and correlates well with their invasion into adjacent tissues and the formation of metastases. Growing evidence indicates that loss of cell-cell adhesion and cell polarity may also be important in early stages of cancer. The strongest hints in this direction come from studies on tumour suppressor genes in the fruitfly Drosophila melanogaster, which have revealed their importance in the control of apical-basal cell polarity.
Collapse
Affiliation(s)
- Andreas Wodarz
- Department of Stem Cell Biology, DFG Research Center for Molecular Physiology of the Brain (CMPB), University of Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany.
| | | |
Collapse
|
92
|
Zhou G, Dada LA, Chandel NS, Iwai K, Lecuona E, Ciechanover A, Sznajder JI. Hypoxia-mediated Na-K-ATPase degradation requires von Hippel Lindau protein. FASEB J 2007; 22:1335-42. [PMID: 18073334 DOI: 10.1096/fj.07-8369com] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hypoxia inhibits Na-K-ATPase activity and leads to its degradation in mammalian cells. Von Hippel Lindau protein (pVHL) and hypoxia inducible factor (HIF) are key mediators in cellular adaptation to hypoxia; thus, we set out to investigate whether pVHL and HIF participate in the hypoxia-mediated degradation of plasma membrane Na-K-ATPase. We found that in the presence of pVHL hypoxia decreased Na-K-ATPase activity and promoted the degradation of plasma membrane Na-K-ATPase. In pVHL-deficient cells, hypoxia did not decrease the Na-K-ATPase activity and the degradation of plasma membrane Na-K-ATPase was prevented. pVHL-mediated degradation of Na-K-ATPase required the functional pVHL E3 ligase and Ubc5 since pVHL mutants and dominant-negative Ubc5 prevented Na-K-ATPase from degradation. The generation of reactive oxygen species was necessary for pVHL-mediated Na-K-ATPase degradation during hypoxia. Desferrioxamine, which stabilizes HIF1/2alpha, did not affect the half-life of plasma membrane Na-K-ATPase. In addition, stabilizing HIF1/2alpha by infecting mammalian cells with adenoviruses containing the oxygen-dependent degradation domain of HIF1alpha did not affect the plasma membrane Na-K-ATPase degradation. In cells with suppression of pVHL by short hairpin RNA, the Na-K-ATPase was not degraded during hypoxia, whereas cells with knockdown of HIF1/2alpha retained the ability to degrade plasma membrane Na-K-ATPase. These findings suggest that pVHL participates in the hypoxia-mediated degradation of plasma membrane Na-K-ATPase in a HIF-independent manner.
Collapse
Affiliation(s)
- Guofei Zhou
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, 240 E. Huron, Chicago, IL 60611, USA
| | | | | | | | | | | | | |
Collapse
|
93
|
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
|
94
|
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
|
95
|
Iturrioz X, Parker PJ. PKCzetaII is a target for degradation through the tumour suppressor protein pVHL. FEBS Lett 2007; 581:1397-402. [PMID: 17350623 DOI: 10.1016/j.febslet.2007.02.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 02/18/2007] [Accepted: 02/23/2007] [Indexed: 10/23/2022]
Abstract
PKCzetaII is a rapidly degraded variant of PKCzeta that suppresses epithelial cell polarisation. It is shown here that PKCzetaII is a target for the E3 ligase and tumour suppressor Von Hippel-Lindau protein (pVHL). Deletion studies demonstrate that the C-terminal region is required for the pVHL and proteasome dependent turnover of PKCzetaII, however it is the N-terminal PB1 domain of PKCzetaII that is required for pVHL complex formation. Reciprocal deletion studies define the pVHL effector domain as the dominant PKCzetaII binding site. The results indicate that pVHL recruits PKCzetaII via its PB1 domain and causes ubiquitination and degradation via the distal C-terminus of PKCzetaII.
Collapse
Affiliation(s)
- Xavier Iturrioz
- Protein Phosphorylation Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields Laboratories, London WC2A 3PX, UK
| | | |
Collapse
|
96
|
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
|
97
|
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
|
98
|
Abstract
Inactivation of the von Hippel-Lindau (VHL) tumor suppressor has been linked to a variety of tumors, including clear cell renal carcinoma, retinal and cerebellar hemangioblastoma, and pheochromocytoma. The best documented function of VHL protein (pVHL) relates to its ability to target the hypoxia-inducible transcription factor (HIF) for polyubiquitylation and proteasomal degradation. This chapter focuses on studies published over the past 2 years related to pVHL. These studies include those describing genetically engineered mice that were used to interrogate the relationship between pVHL and HIF in vivo and cell culture studies that underscore the importance of pVHL in epithelial differentiation and maintenance of the primary cilium. In addition, recent work suggests that pVHL regulates neuronal apoptosis in an HIF-independent manner, and this activity is linked to the risk of developing pheochromocytoma.
Collapse
Affiliation(s)
- William G Kaelin
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
99
|
Nakamura M, Tokunaga F, Sakata SI, Iwai K. Mutual regulation of conventional protein kinase C and a ubiquitin ligase complex. Biochem Biophys Res Commun 2006; 351:340-7. [PMID: 17069764 DOI: 10.1016/j.bbrc.2006.09.163] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Accepted: 09/30/2006] [Indexed: 12/13/2022]
Abstract
Several isoforms of protein kinase C (PKC) are degraded by the ubiquitin-proteasome pathway after phorbol ester-mediated activation. However, little is known about the ubiquitin ligase (E3) that targets activated PKCs. We recently showed that an E3 complex composed of HOIL-1L and HOIP (LUBAC) generates linear polyubiquitin chains and induces the proteasomal degradation of a model substrate. HOIL-1L has also been characterized as a PKC-binding protein. Here we show that LUBAC preferentially binds activated conventional PKCs and their constitutively active mutants. LUBAC efficiently ubiquitinated activated PKC in vitro, and degradation of activated PKCalpha was delayed in HOIL-1L-deficient cells. Conversely, PKC activation induced cleavage of HOIL-1L and led to downregulation of the ligase activity of LUBAC. These results indicate that LUBAC is an E3 for activated conventional PKC, and that PKC and LUBAC regulate each other for proper PKC signaling.
Collapse
Affiliation(s)
- Munehiro Nakamura
- Department of Molecular Cell Biology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | | | | | | |
Collapse
|
100
|
Imanaka M, Iida K, Takahashi K, Tsuji K, Nishizawa H, Fukuoka H, Takeno R, Takahashi Y, Okimura Y, Kaji H, Chihara K. The N131S mutation in the von Hippel-Lindau gene in a Japanese family with pheochromocytoma and hemangioblastomas. Endocr J 2006; 53:819-27. [PMID: 17001110 DOI: 10.1507/endocrj.k06-046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
von Hippel-Lindau (VHL) disease (VHLD) is a hereditary autosomal dominant syndrome that causes various benign and malignant tumors. VHLD is caused by mutations in the VHL tumor suppressor gene. Here, we report a mutation in the VHL gene in a Japanese family with VHLD type 2A, characterized by pheochromocytoma (PHE), and hemangioblastomas (HAB) in both the retina and thoracic spinal cord but without renal cell carcinoma (RCC). We identified a heterozygous A to G point mutation at the second base of codon 131 of the VHL protein (pVHL). This mutation was predicted to convert codon 131 from asparagine to serine (N131S). Although most mutations in VHLD type 2A have been detected in the alpha domain of pVHL, the present mutated amino acid was located at the region encoding the beta domain of pVHL. Previous patients with the N131K or N131T mutation in pVHL developed VHLD type 2B with RCC or VHLD type 1 without PHE, respectively. We also identified somatic loss of heterozygosity (LOH) at chromosome 3p25-26 in the adrenal tumor of the patient. The results of our study suggest that not only the location of mutation but also the altered amino acid may be critical for determining the clinical phenotype of VHLD. LOH was associated with the development of PHE in a patient with the N131S mutation in pVHL.
Collapse
Affiliation(s)
- Mari Imanaka
- Division of Endocrinology/Metabolism, Neurology, and Hematology/Oncology, Department of Clinical Molecular Medicine, Kobe University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|