1
|
Morriss S, Beshay V, Leong HS, Winship I. Novel Case of Bilateral Adrenal Tumors Confirms Pathogenicity of Previously Described c.463+4C>G Variant in the von-Hippel Lindau Gene. J Kidney Cancer VHL 2025; 12:23-26. [PMID: 40051608 PMCID: PMC11884336 DOI: 10.15586/jkc.v12i1.381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
We report a case of a pathogenic variant c.463+4C>G in the von Hippel-Lindau (VHL) gene identified in a patient presenting with bilateral adrenal tumors, including a histologically confirmed pheochromocytoma with no significant family history of VHL-associated tumors. This same variant was first reported as having pathogenic significance in an unrelated proband with a hemangioblastoma and a family history of pheochromocytoma. In our patient, next-generation sequencing and subsequent RNA (ribonucleic acid) analysis confirmed this mutation to be a pathogenic (class 4) variant in intron 2. The lack of family history of VHL-associated tumors correlated with the proband further suggests that this mutation may have reduced penetrance. This case confirms the pathogenicity of the same previously described variant in the VHL gene and underscores the utility of genetic testing in patients with atypical presentations of adrenal tumors, even in the absence of a relevant family history.
Collapse
Affiliation(s)
- Samuel Morriss
- Familial Cancer Centre, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Victoria Beshay
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Huei San Leong
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ingrid Winship
- Familial Cancer Centre, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Carlton, Victoria, Australia
| |
Collapse
|
2
|
Duan E, Robinson M, Davis C, Pruthi S, Shin C, Lewis M, Martinez-Agosto JA, Gorin MB, Shuch BM, Friedman DL, Chang VY. Pediatric patients with von Hippel-Lindau and hemangioblastomas treated successfully with belzutifan. Pediatr Blood Cancer 2025; 72:e31371. [PMID: 39415342 DOI: 10.1002/pbc.31371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/24/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024]
Abstract
Hemangioblastoma is the most common tumor associated with von Hippel-Lindau (VHL), and are a leading cause of mortality. We present five pediatric patients with VHL-associated hemangioblastomas treated with belzutifan, a hypoxia-inducible factor 2a (HIF2a) inhibitor. Three patients were started on belzutifan due to vision loss from progressive retinal hemangioblastomas. Within one year of treatment, all three patients had improvement in hemangioblastoma size and visual acuity. For patients with intracranial lesions, belzutifan resulted in an improvement in neurologic symptoms and hemangioblastoma size. Four patients experienced grade 1-2 anemia and two patients required a dose reduction. Our report suggests that belzutifan can be an effective therapy for pediatric, adolescent, and young adult patients with VHL-associated hemangioblastomas.
Collapse
Affiliation(s)
- Emily Duan
- Department of Pediatrics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Michael Robinson
- Division of Pediatric Hematology/Oncology, Vanderbilt, Nashville, Tennessee, USA
| | - Charles Davis
- Department of Radiology, UCLA, Los Angeles, California, USA
| | - Sumit Pruthi
- Department of Neuroradiology, Vanderbilt, Nashville, Tennessee, USA
| | - Christina Shin
- Ronald Reagan UCLA Medical Center, UCLA, Los Angeles, California, USA
| | - Marisa Lewis
- Division of Pediatric Hematology/Oncology, UCLA, Los Angeles, California, USA
| | - Julian A Martinez-Agosto
- Division of Clinical Genetics, Department of Human Genetics, UCLA, Los Angeles, California, USA
- Department of Psychiatry, UCLA, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Michael B Gorin
- Department of Ophthalmology, UCLA, Los Angeles, California, USA
| | - Brian M Shuch
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
- Department of Urology, UCLA, Los Angeles, California, USA
| | - Debra L Friedman
- Division of Pediatric Hematology/Oncology, Vanderbilt, Nashville, Tennessee, USA
| | - Vivian Y Chang
- Division of Pediatric Hematology/Oncology, UCLA, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
- Children's Discovery and Innovation Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|
3
|
Cicchetti R, Basconi M, Litterio G, Mascitti M, Tamborino F, Orsini A, Digiacomo A, Ferro M, Schips L, Marchioni M. Advances in Molecular Mechanisms of Kidney Disease: Integrating Renal Tumorigenesis of Hereditary Cancer Syndrome. Int J Mol Sci 2024; 25:9060. [PMID: 39201746 PMCID: PMC11355026 DOI: 10.3390/ijms25169060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Renal cell carcinoma (RCC) comprises various histologically distinct subtypes, each characterized by specific genetic alterations, necessitating individualized management and treatment strategies for each subtype. An exhaustive search of the PubMed database was conducted without any filters or restrictions. Inclusion criteria encompassed original English articles focusing on molecular mechanisms of kidney cancer. On the other hand, all non-original articles and articles published in any language other than English were excluded. Hereditary kidney cancer represents 5-8% of all kidney cancer cases and is associated with syndromes such as von Hippel-Lindau syndrome, Birt-Hogg-Dubè syndrome, succinate dehydrogenase-deficient renal cell cancer syndrome, tuberous sclerosis complex, hereditary papillary renal cell carcinoma, fumarate hydratase deficiency syndrome, BAP1 tumor predisposition syndrome, and other uncommon hereditary cancer syndromes. These conditions are characterized by distinct genetic mutations and related extra-renal symptoms. The majority of renal cell carcinoma predispositions stem from loss-of-function mutations in tumor suppressor genes. These mutations promote malignant advancement through the somatic inactivation of the remaining allele. This review aims to elucidate the main molecular mechanisms underlying the pathophysiology of major syndromes associated with renal cell carcinoma. By providing a comprehensive overview, it aims to facilitate early diagnosis and to highlight the principal therapeutic options available.
Collapse
Affiliation(s)
- Rossella Cicchetti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Martina Basconi
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Giulio Litterio
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Marco Mascitti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Flavia Tamborino
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Angelo Orsini
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Alessio Digiacomo
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20141 Milan, Italy;
| | - Luigi Schips
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Michele Marchioni
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| |
Collapse
|
4
|
Mukhopadhyay D, Chakraborty B, Sarkar S, Alam N, Panda CK. Clinical implications of activation of the LIMD1-VHL-HIF1α pathway during head-&-neck squamous cell carcinoma development. Indian J Med Res 2024; 159:479-493. [PMID: 39382421 PMCID: PMC11463245 DOI: 10.25259/ijmr_1262_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Indexed: 10/10/2024] Open
Abstract
Background & objectives Given the importance of the role of hypoxia induced pathway in different cancers including head-and-neck squamous cell carcinoma (HNSCC), this study delved into elucidating the molecular mechanism of hypoxia-inducible factor-1α (HIF1α) activation in HNSCC. Additionally, it analyzes the alterations of its regulatory genes [von Hippel-Lindau (VHL) and LIM domain containing 1 (LIMD1)] and target gene vascular endothelial growth factor (VEGF) in head-and-neck lesions at different clinical stages in relation with human papillomavirus (HPV) infection. Methods Global mRNA expression profiles of HIF1α, VHL, LIMD1 and VEGF were evaluated from public datasets of HNSCC, followed by validation of their expression (mRNA/protein) in an independent set of HPV+ve/-ve HNSCC samples of different clinical stages. Results A diverse expression pattern of the HIF1α pathway genes was observed, irrespective of HPV infection, in the datasets. In validation in an independent set of HNSCC samples, high mRNA expressions of HIF1α/VEGF were observed particularly in HPV positive samples. However, VHL/LIMD1 mRNA expression was low in tumours regardless of HPV infection status. In immunohistochemical analysis, high/medium (H/M) expression of HIF1α/VEGF was observed in basal/parabasal layers of normal epithelium, with significantly higher expression in tumours, especially in HPV-positive samples. Conversely, high cytoplasmic VHL expression in these layers gradually decreased with the progression of HNSCC, regardless of HPV infection. A similar trend was noted in LIMD1 expression (nuclear/cytoplasmic) during the disease development. The methylation pattern of VHL and LIMD1 promoters in the basal/parabasal layers of normal epithelium correlated with their expression, exhibiting a gradual increase with the progression of HNSCC. The H/M expression of HIF1α/VEGF proteins and reduced VHL expression was associated with poor clinical outcomes. Interpretation & conclusions The results of this study showed differential regulation of the LIMD1-VHL-HIF1α pathway in HPV positive and negative HNSCC samples, illustrating the molecular distinctiveness of these two groups.
Collapse
Affiliation(s)
- Debalina Mukhopadhyay
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Balarko Chakraborty
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Shreya Sarkar
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
- New Brunswick Heart Centre, Saint John, NB, Canada
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| |
Collapse
|
5
|
Kumar V, Kaushik V, Kumar S, Levkovich SA, Gupta P, Laor Bar-Yosef D, Gazit E, Segal D. The von Hippel-Lindau protein forms fibrillar amyloid assemblies that are mitigated by the anti-amyloid molecule Purpurin. Biochem Biophys Res Commun 2024; 690:149250. [PMID: 38039781 DOI: 10.1016/j.bbrc.2023.149250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023]
Abstract
The von Hippel-Lindau protein (pVHL) is a tumor suppressor involved in oxygen regulation via dynamic nucleocytoplasmic shuttling. It plays a crucial role in cell survival by degrading hypoxia-inducible factors (HIFs). Mutations in the VHL gene cause angiogenic tumors, characterized as VHL syndrome. However, aggressive tumors involving wild-type pVHL have also been described but the underlying mechanism remains to be revealed. We have previously shown that pVHL possesses several short amyloid-forming motifs, making it aggregation-prone. In this study, using a series of biophysical assays, we demonstrated that a pVHL-derived fragment (pVHL104-140) that harbors the nuclear export motif and HIF binding site, forms amyloid-like fibrillar structures in vitro by following secondary-nucleation-based kinetics. The peptide also formed amyloids at acidic pH that mimics the tumor microenvironment. We, subsequently, validated the amyloid formation by pVHL in vitro. Using the Curli-dependent amyloid generator (C-DAG) expression system, we confirmed the amyloidogenesis of pVHL in bacterial cells. The pVHL amyloids are an attractive target for therapeutics of the VHL syndrome. Accordingly, we demonstrated in vitro that Purpurin is a potent inhibitor of pVHL fibrillation. The amyloidogenic behavior of wild-type pVHL and its inhibition provide novel insights into the molecular underpinning of the VHL syndrome and its possible treatment.
Collapse
Affiliation(s)
- Vijay Kumar
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Vibha Kaushik
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Sourav Kumar
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Shon A Levkovich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Priya Gupta
- School of Plant Sciences and Food Security, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Dana Laor Bar-Yosef
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Ehud Gazit
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel; BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Daniel Segal
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel; BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
6
|
Kanno H, Matsumoto S, Yoshizumi T, Nakahara K, Kubo A, Murata H, Shuin T, U HS. Role of SOCS and VHL Proteins in Neuronal Differentiation and Development. Int J Mol Sci 2023; 24:ijms24043880. [PMID: 36835292 PMCID: PMC9960776 DOI: 10.3390/ijms24043880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
The basic helix-loop-helix factors play a central role in neuronal differentiation and nervous system development, which involve the Notch and signal transducer and activator of transcription (STAT)/small mother against decapentaplegic signaling pathways. Neural stem cells differentiate into three nervous system lineages, and the suppressor of cytokine signaling (SOCS) and von Hippel-Lindau (VHL) proteins are involved in this neuronal differentiation. The SOCS and VHL proteins both contain homologous structures comprising the BC-box motif. SOCSs recruit Elongin C, Elongin B, Cullin5(Cul5), and Rbx2, whereas VHL recruits Elongin C, Elongin B, Cul2, and Rbx1. SOCSs form SBC-Cul5/E3 complexes, and VHL forms a VBC-Cul2/E3 complex. These complexes degrade the target protein and suppress its downstream transduction pathway by acting as E3 ligases via the ubiquitin-proteasome system. The Janus kinase (JAK) is the main target protein of the E3 ligase SBC-Cul5, whereas hypoxia-inducible factor is the primary target protein of the E3 ligase VBC-Cul2; nonetheless, VBC-Cul2 also targets the JAK. SOCSs not only act on the ubiquitin-proteasome system but also act directly on JAKs to suppress the Janus kinase-signal transduction and activator of transcription (JAK-STAT) pathway. Both SOCS and VHL are expressed in the nervous system, predominantly in brain neurons in the embryonic stage. Both SOCS and VHL induce neuronal differentiation. SOCS is involved in differentiation into neurons, whereas VHL is involved in differentiation into neurons and oligodendrocytes; both proteins promote neurite outgrowth. It has also been suggested that the inactivation of these proteins may lead to the development of nervous system malignancies and that these proteins may function as tumor suppressors. The mechanism of action of SOCS and VHL involved in neuronal differentiation and nervous system development is thought to be mediated through the inhibition of downstream signaling pathways, JAK-STAT, and hypoxia-inducible factor-vascular endothelial growth factor pathways. In addition, because SOCS and VHL promote nerve regeneration, they are expected to be applied in neuronal regenerative medicine for traumatic brain injury and stroke.
Collapse
Affiliation(s)
- Hiroshi Kanno
- Department of Neurosurgery, School of Medicine, Yokohama City University, Yokohama 232-0024, Japan
- Department of Neurosurgery, Asahi Hospital, Tokyo 121-0078, Japan
- Correspondence: ; Tel.: +81-3-5242-5800
| | - Shutaro Matsumoto
- Department of Neurosurgery, School of Medicine, Yokohama City University, Yokohama 232-0024, Japan
- Department of Neurosurgery, Asahi Hospital, Tokyo 121-0078, Japan
| | - Tetsuya Yoshizumi
- Department of Neurosurgery, St. Mariannna Medical University, Kawasaki 216-8511, Japan
| | - Kimihiro Nakahara
- Department of Neurosurgery, International University of Health and Welfare, Atami 413-0012, Japan
| | | | - Hidetoshi Murata
- Department of Neurosurgery, St. Mariannna Medical University, Kawasaki 216-8511, Japan
| | - Taro Shuin
- Kochi Medical School Hospital, Nangoku 783-0043, Japan
| | - Hoi-Sang U
- Department of Electrical Engineering, University of California San Diego, San Diego, CA 92093, USA
| |
Collapse
|
7
|
Loss of sphingosine kinase 2 promotes the expansion of hematopoietic stem cells by improving their metabolic fitness. Blood 2022; 140:1686-1701. [PMID: 35881840 DOI: 10.1182/blood.2022016112] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have reduced capacities to properly maintain and replenish the hematopoietic system during myelosuppressive injury or aging. Expanding and rejuvenating HSCs for therapeutic purposes has been a long-sought goal with limited progress. Here, we show that the enzyme Sphk2 (sphingosine kinase 2), which generates the lipid metabolite sphingosine-1-phosphate, is highly expressed in HSCs. The deletion of Sphk2 markedly promotes self-renewal and increases the regenerative potential of HSCs. More importantly, Sphk2 deletion globally preserves the young HSC gene expression pattern, improves the function, and sustains the multilineage potential of HSCs during aging. Mechanistically, Sphk2 interacts with prolyl hydroxylase 2 and the Von Hippel-Lindau protein to facilitate HIF1α ubiquitination in the nucleus independent of the Sphk2 catalytic activity. Deletion of Sphk2 increases hypoxic responses by stabilizing the HIF1α protein to upregulate PDK3, a glycolysis checkpoint protein for HSC quiescence, which subsequently enhances the function of HSCs by improving their metabolic fitness; specifically, it enhances anaerobic glycolysis but suppresses mitochondrial oxidative phosphorylation and generation of reactive oxygen species. Overall, targeting Sphk2 to enhance the metabolic fitness of HSCs is a promising strategy to expand and rejuvenate functional HSCs.
Collapse
|
8
|
Fefilova AS, Antifeeva IA, Gavrilova AA, Turoverov KK, Kuznetsova IM, Fonin AV. Reorganization of Cell Compartmentalization Induced by Stress. Biomolecules 2022; 12:1441. [PMID: 36291650 PMCID: PMC9599104 DOI: 10.3390/biom12101441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/17/2022] Open
Abstract
The discovery of intrinsically disordered proteins (IDPs) that do not have an ordered structure and nevertheless perform essential functions has opened a new era in the understanding of cellular compartmentalization. It threw the bridge from the mostly mechanistic model of the organization of the living matter to the idea of highly dynamic and functional "soft matter". This paradigm is based on the notion of the major role of liquid-liquid phase separation (LLPS) of biopolymers in the spatial-temporal organization of intracellular space. The LLPS leads to the formation of self-assembled membrane-less organelles (MLOs). MLOs are multicomponent and multifunctional biological condensates, highly dynamic in structure and composition, that allow them to fine-tune the regulation of various intracellular processes. IDPs play a central role in the assembly and functioning of MLOs. The LLPS importance for the regulation of chemical reactions inside the cell is clearly illustrated by the reorganization of the intracellular space during stress response. As a reaction to various types of stresses, stress-induced MLOs appear in the cell, enabling the preservation of the genetic and protein material during unfavourable conditions. In addition, stress causes structural, functional, and compositional changes in the MLOs permanently present inside the cells. In this review, we describe the assembly of stress-induced MLOs and the stress-induced modification of existing MLOs in eukaryotes, yeasts, and prokaryotes in response to various stress factors.
Collapse
Affiliation(s)
| | | | | | - Konstantin K. Turoverov
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of RAS, 194064 St. Petersburg, Russia
| | | | | |
Collapse
|
9
|
Abstract
Germline loss-of-function mutations of the VHL tumor suppressor gene cause von Hippel–Lindau disease, which is associated with an increased risk of hemangioblastomas, clear cell renal cell carcinomas (ccRCCs), and paragangliomas. This Review describes mechanisms involving the VHL gene product in oxygen sensing, protein degradation, and tumor development and current therapeutic strategies targeting these mechanisms. The VHL gene product is the substrate recognition subunit of a ubiquitin ligase that targets the α subunit of the heterodimeric hypoxia-inducible factor (HIF) transcription factor for proteasomal degradation when oxygen is present. This oxygen dependence stems from the requirement that HIFα be prolyl-hydroxylated on one (or both) of two conserved prolyl residues by members of the EglN (also called PHD) prolyl hydroxylase family. Deregulation of HIF, and particularly HIF2, drives the growth of VHL-defective ccRCCs. Drugs that inhibit the HIF-responsive gene product VEGF are now mainstays of ccRCC treatment. An allosteric HIF2 inhibitor was recently approved for the treatment of ccRCCs arising in the setting of VHL disease and has advanced to phase III testing for sporadic ccRCCs based on promising phase I/II data. Orally available EglN inhibitors are being tested for the treatment of anemia and ischemia. Five of these agents have been approved for the treatment of anemia in the setting of chronic kidney disease in various countries around the world.
Collapse
|
10
|
Liu X, Zurlo G, Zhang Q. The Roles of Cullin-2 E3 Ubiquitin Ligase Complex in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1217:173-186. [PMID: 31898228 DOI: 10.1007/978-981-15-1025-0_11] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Posttranslational protein modifications play an important role in regulating protein stability and cellular function. There are at least eight Cullin family members. Among them, Cullin-2 forms a functional E3 ligase complex with elongin B, elongin C, RING-box protein 1 (RBX1, also called ROC1), as well as the substrate recognition subunit (SRS) to promote the substrate ubiquitination and degradation. In this book chapter, we will review Cullin-2 E3 ligase complexes that include various SRS proteins, including von Hippel Lindau (pVHL), leucine-rich repeat protein-1 (LRR-1), preferentially expressed antigen of melanoma (PRAME), sex-determining protein FEM-1 and early embryogenesis protein ZYG-11. We will focus on the VHL signaling pathway in clear cell renal cell carcinoma (ccRCC), which may reveal various therapeutic avenues in treating this lethal cancer.
Collapse
Affiliation(s)
- Xijuan Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Giada Zurlo
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA. .,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA. .,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA. .,Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
11
|
Taylor J, Sendino M, Gorelick AN, Pastore A, Chang MT, Penson AV, Gavrila EI, Stewart C, Melnik EM, Herrejon Chavez F, Bitner L, Yoshimi A, Lee SCW, Inoue D, Liu B, Zhang XJ, Mato AR, Dogan A, Kharas MG, Chen Y, Wang D, Soni RK, Hendrickson RC, Prieto G, Rodriguez JA, Taylor BS, Abdel-Wahab O. Altered Nuclear Export Signal Recognition as a Driver of Oncogenesis. Cancer Discov 2019; 9:1452-1467. [PMID: 31285298 PMCID: PMC6774834 DOI: 10.1158/2159-8290.cd-19-0298] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 12/17/2022]
Abstract
Altered expression of XPO1, the main nuclear export receptor in eukaryotic cells, has been observed in cancer, and XPO1 has been a focus of anticancer drug development. However, mechanistic evidence for cancer-specific alterations in XPO1 function is lacking. Here, genomic analysis of 42,793 cancers identified recurrent and previously unrecognized mutational hotspots in XPO1. XPO1 mutations exhibited striking lineage specificity, with enrichment in a variety of B-cell malignancies, and introduction of single amino acid substitutions in XPO1 initiated clonal, B-cell malignancy in vivo. Proteomic characterization identified that mutant XPO1 altered the nucleocytoplasmic distribution of hundreds of proteins in a sequence-specific manner that promoted oncogenesis. XPO1 mutations preferentially sensitized cells to inhibitors of nuclear export, providing a biomarker of response to this family of drugs. These data reveal a new class of oncogenic alteration based on change-of-function mutations in nuclear export signal recognition and identify therapeutic targets based on altered nucleocytoplasmic trafficking. SIGNIFICANCE: Here, we identify that heterozygous mutations in the main nuclear exporter in eukaryotic cells, XPO1, are positively selected in cancer and promote the initiation of clonal B-cell malignancies. XPO1 mutations alter nuclear export signal recognition in a sequence-specific manner and sensitize cells to compounds in clinical development inhibiting XPO1 function.This article is highlighted in the In This Issue feature, p. 1325.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Cell Proliferation
- Cell Transformation, Neoplastic
- Disease Models, Animal
- Gene Expression
- Genes, bcl-2
- Genes, myc
- Humans
- Karyopherins/chemistry
- Karyopherins/genetics
- Karyopherins/metabolism
- Leukemia, B-Cell/genetics
- Leukemia, B-Cell/metabolism
- Leukemia, B-Cell/mortality
- Leukemia, B-Cell/pathology
- Mice
- Mutation
- Nuclear Export Signals
- Organ Specificity/genetics
- Protein Binding
- Receptors, Cytoplasmic and Nuclear/chemistry
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Structure-Activity Relationship
- Exportin 1 Protein
Collapse
Affiliation(s)
- Justin Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria Sendino
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, Barrio Sarriena s/n, Leioa, Spain
| | - Alexander N Gorelick
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alessandro Pastore
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew T Chang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander V Penson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elena I Gavrila
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Connor Stewart
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ella M Melnik
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Lillian Bitner
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Akihide Yoshimi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stanley Chun-Wei Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daichi Inoue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bo Liu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xiao J Zhang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anthony R Mato
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael G Kharas
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuhong Chen
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin
| | - Demin Wang
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin
| | - Rajesh K Soni
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald C Hendrickson
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gorka Prieto
- Department of Communications Engineering, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Jose A Rodriguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, Barrio Sarriena s/n, Leioa, Spain
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
12
|
Florini F, Naguleswaran A, Gharib WH, Bringaud F, Roditi I. Unexpected diversity in eukaryotic transcription revealed by the retrotransposon hotspot family of Trypanosoma brucei. Nucleic Acids Res 2019; 47:1725-1739. [PMID: 30544263 PMCID: PMC6393297 DOI: 10.1093/nar/gky1255] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/20/2022] Open
Abstract
The path from DNA to RNA to protein in eukaryotes is guided by a series of factors linking transcription, mRNA export and translation. Many of these are conserved from yeast to humans. Trypanosomatids, which diverged early in the eukaryotic lineage, exhibit unusual features such as polycistronic transcription and trans-splicing of all messenger RNAs. They possess basal transcription factors, but lack recognisable orthologues of many factors required for transcription elongation and mRNA export. We show that retrotransposon hotspot (RHS) proteins fulfil some of these functions and that their depletion globally impairs nascent RNA synthesis by RNA polymerase II. Three sub-families are part of a coordinated process in which RHS6 is most closely associated with chromatin, RHS4 is part of the Pol II complex and RHS2 connects transcription with the translation machinery. In summary, our results show that the components of eukaryotic transcription are far from being universal, and reveal unsuspected plasticity in the course of evolution.
Collapse
Affiliation(s)
- Francesca Florini
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Graduate School of Cellular and Biomedical Science, University of Bern, Bern, Switzerland
| | | | - Walid H Gharib
- Interfaculty Bioinformatics Unit, University of Bern, Switzerland
| | - Frédéric Bringaud
- Laboratoire de Microbiologie Fondamentale et Pathogénicité (MFP), UMR 5234 CNRS, Université de Bordeaux, France
| | - Isabel Roditi
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Fu C, Tyagi R, Chin AC, Rojas T, Li RJ, Guha P, Bernstein IA, Rao F, Xu R, Cha JY, Xu J, Snowman AM, Semenza GL, Snyder SH. Inositol Polyphosphate Multikinase Inhibits Angiogenesis via Inositol Pentakisphosphate-Induced HIF-1α Degradation. Circ Res 2017; 122:457-472. [PMID: 29279301 DOI: 10.1161/circresaha.117.311983] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022]
Abstract
RATIONALE Inositol polyphosphate multikinase (IPMK) and its major product inositol pentakisphosphate (IP5) regulate a variety of cellular functions, but their role in vascular biology remains unexplored. OBJECTIVE We have investigated the role of IPMK in regulating angiogenesis. METHODS AND RESULTS Deletion of IPMK in fibroblasts induces angiogenesis in both in vitro and in vivo models. IPMK deletion elicits a substantial increase of VEGF (vascular endothelial growth factor), which mediates the regulation of angiogenesis by IPMK. The regulation of VEGF by IPMK requires its catalytic activity. IPMK is predominantly nuclear and regulates gene transcription. However, IPMK does not apparently serve as a transcription factor for VEGF. HIF (hypoxia-inducible factor)-1α is a major determinant of angiogenesis and induces VEGF transcription. IPMK deletion elicits a major enrichment of HIF-1α protein and thus VEGF. HIF-1α is constitutively ubiquitinated by pVHL (von Hippel-Lindau protein) followed by proteasomal degradation under normal conditions. However, HIF-1α is not recognized and ubiquitinated by pVHL in IPMK KO (knockout) cells. IP5 reinstates the interaction of HIF-1α and pVHL. HIF-1α prolyl hydroxylation, which is prerequisite for pVHL recognition, is interrupted in IPMK-deleted cells. IP5 promotes HIF-1α prolyl hydroxylation and thus pVHL-dependent degradation of HIF-1α. Deletion of IPMK in mouse brain increases HIF-1α/VEGF levels and vascularization. The increased VEGF in IPMK KO disrupts blood-brain barrier and enhances brain blood vessel permeability. CONCLUSIONS IPMK, via its product IP5, negatively regulates angiogenesis by inhibiting VEGF expression. IP5 acts by enhancing HIF-1α hydroxylation and thus pVHL-dependent degradation of HIF-1α.
Collapse
Affiliation(s)
- Chenglai Fu
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richa Tyagi
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alfred C Chin
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tomas Rojas
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ruo-Jing Li
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Prasun Guha
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Isaac A Bernstein
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Feng Rao
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Risheng Xu
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jiyoung Y Cha
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jing Xu
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Adele M Snowman
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gregg L Semenza
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Solomon H Snyder
- From the Solomon H. Snyder Department of Neuroscience (C.F., R.T., A.C.C., T.R., P.G., I.A.B., F.R., R.X., J.Y.C., J.X., A.M.S., S.H.S.), Department of Pharmacology and Molecular Sciences (R.-J.L., S.H.S.), Institute for Cell Engineering (G.L.S.), McKusick-Nathans Institute of Genetic Medicine (G.L.S.), Department of Pediatrics (G.L.S.), Department of Medicine (G.L.S.), Department of Oncology (G.L.S.), Department of Radiation Oncology (G.L.S.), Department of Biological Chemistry (G.L.S.), and Department of Psychiatry and Behavioral Sciences (S.H.S.), Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
14
|
KAELIN WILLIAMG. The VHL Tumor Suppressor Gene: Insights into Oxygen Sensing and Cancer. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2017; 128:298-307. [PMID: 28790514 PMCID: PMC5525432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Mammalian cells sense changes in oxygen and transduce that information into adaptive changes in gene expression using a conserved pathway that converges on the heterodimeric transcription factor called hypoxia-inducible factor (HIF), which contains a labile alpha subunit and a stable beta subunit. In the presence of oxygen, the alpha subunit is hydroxylated on one (or both) of two highly conserved prolyl residues by an Egg-Laying Defective Nine (EglN) [also called Prolyl Hydroxylase Domain (PHD)] dioxygenase, which recruits an ubiquitin ligase complex containing the VHL tumor suppressor gene product. Germline VHL mutations cause von Hippel-Lindau (VHL) disease, which manifest as angiogenic tumors such as hemangioblastomas and kidney cancers. Somatic VHL inactivation and deregulation of HIF (especially HIF2α) drives sporadic kidney cancers and an HIF2α inhibitor is showing promise for this disease. VHL, EglN1, and HIF2α polymorphisms have been linked to familial polycythemia and adaptation to high altitude. Orally available EglN inhibitors are being developed for the treatment of anemia and ischemic diseases.
Collapse
Affiliation(s)
- WILLIAM G. KAELIN
- Correspondence and reprint requests: William G. Kaelin, Jr., MD,
450 Brookline Ave., Mayer 457, Boston, Massachusetts 02215617-632-3975617-632-4760
| |
Collapse
|
15
|
Vivek PJ, Resmi MS, Sreekumar S, Sivakumar KC, Tuteja N, Soniya EV. Calcium-Dependent Protein Kinase in Ginger Binds with Importin-α through Its Junction Domain for Nuclear Localization, and Further Interacts with NAC Transcription Factor. FRONTIERS IN PLANT SCIENCE 2016; 7:1909. [PMID: 28133460 PMCID: PMC5233720 DOI: 10.3389/fpls.2016.01909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/02/2016] [Indexed: 05/11/2023]
Abstract
Calcium-dependent protein kinases (CDPKs) are important sensors of Ca2+ elevations in plant cells regulating the gene expression linked with various cellular processes like stress response, growth and development, metabolism, and cytoskeleton dynamics. Ginger is an extensively used spice due to its unique flavor and immense medicinal value. The two major threats that interfere with the large scale production of ginger are the salinity and drought stress. ZoCDPK1 (Zingiber officinale Calcium-dependent protein kinase 1) is a salinity and drought-inducible CDPK gene isolated from ginger and undergoes dynamic subcellular localization during stress conditions. ZoCDPK1, with signature features of a typical Ca2+ regulated kinase, also possesses a bipartite nuclear localization sequence (NLS) in its junction domain (JD). A striking feature in ZoCDPK1 is the rare occurrence of a coupling between the NLS in JD and consensus sequences in regulatory domain. Here, we further identified its nature of nuclear localization and its interaction partners. In the homology model generated for ZoCDPK1, the regulatory domain mimics the crystal structure of the regulatory domain in Arabidopsis CDPK1. Molecular docking simulation of importin (ZoIMPα), an important protein involved in nuclear translocation, into the NLS of ZoCDPK1 was well-visualized. Furthermore, the direct interaction of ZoCDPK1 and ZoIMPα proteins was studied by the yeast 2-hybrid (Y2H) system, which confirmed that junction domain (JD) is an important interaction module required for ZoCDPK1 and ZoIMPα binding. The probable interacting partners of ZoCDPK1 were also identified using Y2H experiment. Of the 10 different stress-related interacting partners identified for ZoCDPK1, NAC transcription factor (TF) needs special mention, especially in the context of ZoCDPK1 function. The interaction between ZoCDPK1 and NAC TF, in fact, corroborate with the results of gene expression and over-expression studies of ZoCDPK1. Hence ZoCDPK1 is operating through NAC TF mediated ABA-independent, cold non-responsive stress signaling pathway in ginger.
Collapse
Affiliation(s)
| | | | - Sweda Sreekumar
- Rajiv Gandhi Centre for BiotechnologyThiruvananthapuram, India
| | - K. C. Sivakumar
- Rajiv Gandhi Centre for BiotechnologyThiruvananthapuram, India
| | - Narendra Tuteja
- Amity Institute of Microbial Technology, Amity UniversityNoida, India
| | - Eppurathu Vasudevan Soniya
- Rajiv Gandhi Centre for BiotechnologyThiruvananthapuram, India
- *Correspondence: Eppurathu Vasudevan Soniya
| |
Collapse
|
16
|
Jiang X, Zhang D, Zhang H, Huang Y, Teng M. Role of Ran-regulated nuclear-cytoplasmic trafficking of pVHL in the regulation of microtubular stability-mediated HIF-1α in hypoxic cardiomyocytes. Sci Rep 2015; 5:9193. [PMID: 25779090 PMCID: PMC4361876 DOI: 10.1038/srep09193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 02/25/2015] [Indexed: 12/30/2022] Open
Abstract
Our previous study suggested that microtubule network alteration affects the process of glycolysis in cardiomyocytes (CMs) via the regulation of hypoxia-inducible factor (HIF)-1α during the early stages of hypoxia. However, little is known regarding the underlying mechanisms of microtubule network alteration-induced changes of HIF-1α. The von Hippel–Lindau tumor suppressor protein (pVHL) has been shown to mediate the ubiquitination of HIF-1α in the nuclear compartment prior to HIF-1α exportation to the cytoplasm, and pVHL dynamic nuclear-cytoplasmic trafficking is indicated to be involved in the process of HIF-1α degradation. In this study, by administering different microtubule-stabilizing and -depolymerizing interventions, we demonstrated that microtubule stabilization promoted pVHL nuclear export and drove the translocation of pVHL to the cytoplasm, while microtubule disruption prevented pVHL nuclear export in hypoxic CMs. Moreover, the ratio between nuclear and cytoplasmic pVHL was associated with HIF-1α regulation. Importantly, microtubule network alteration also affected the subcellular localization of Ran, which was involved in the regulation of pVHL nuclear-cytoplasmic trafficking. The above results suggest that the subcellular translocation of pVHL plays an important role in microtubular structure alteration-induced HIF-1α regulation. Interestingly, Ran is involved in the process of pVHL nuclear-cytoplasmic trafficking following microtubule network alteration in hypoxic CMs.
Collapse
Affiliation(s)
- Xupin Jiang
- 1] Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing, China [2] Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dongxia Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Hengshu Zhang
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Miao Teng
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
|
18
|
Abstract
von Hippel-Lindau (VHL) disease is an inheritable condition with an incidence of 1 in 36000 live births. Individuals with VHL develop benign and malignant tumors including retinal and central nervous system hemangioblastomas, clear cell renal cell carcinomas (RCC), pheochromocytomas, pancreatic neuroendocrine tumors and endolymphatic sac tumors (ELSTs). VHL is caused by germline loss of function of the VHL gene on one allele at chromosome 3p25-26. A somatic "second hit" event leads to the loss of the other allele and tumor formation. Loss of VHL function in cells leads to increased expression and stabilization of hypoxia inducible factor (HIF). VHL protein/HIF pathway has been implicated in tumorigenesis for hemangioblastomas, RCC and other VHL tumors. Clinical examination, imaging, and genetic testing for VHL mutations confirm VHL disease. Management of VHL disease largely consists of surgical resection of symptomatic tumors (hemangioblastomas), tumors prone to metastasize (RCC larger than 3cm), or tumors causing hormonal symptoms (pheochromocytomas). Despite advances in early diagnosis and management of VHL disease, life expectancy for VHL patients remains low at 40-52 years. Secondary effects from VHL manifestations are mitigated by routine surveillance and early detection. In this chapter, we summarize the current state of knowledge in VHL disease.
Collapse
Affiliation(s)
- Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Russell R Lonser
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Department of Neurological Surgery, Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
19
|
Role of compartmentalization on HiF-1α degradation dynamics during changing oxygen conditions: a computational approach. PLoS One 2014; 9:e110495. [PMID: 25338163 PMCID: PMC4206521 DOI: 10.1371/journal.pone.0110495] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 09/21/2014] [Indexed: 12/25/2022] Open
Abstract
HiF-1α is the central protein driving the cellular response to hypoxia. Its accumulation in cancer cells is linked to the appearance of chemoresistant and aggressive tumor phenotypes. As a consequence, understanding the regulation of HiF-1α dynamics is a major issue to design new anti-cancer therapies. In this paper, we propose a model of the hypoxia pathway, involving HiF-1α and its inhibitor pVHL. Based on data from the literature, we made the hypothesis that the regulation of HiF-1α involves two compartments (nucleus and cytoplasm) and a constitutive shuttle of the pVHL protein between them. We first show that this model captures correctly the main features of HiF-1α dynamics, including the bi-exponential degradation profile in normoxia, the kinetics of induction in hypoxia, and the switch-like accumulation. Second, we simulated the effects of a hypoxia/reoxygenation event, and show that it generates a strong instability of HiF-1α. The protein concentration rapidly increases 3 hours after the reoxygenation, and exhibits an oscillating pattern. This effect vanishes if we do not consider compartmentalization of HiF-1α. This result can explain various counter-intuitive observations about the specific molecular and cellular response to the reoxygenation process. Third, we simulated the HiF-1α dynamics in the tumor case. We considered different types of mutations associated with tumorigenesis, and we compared their consequences on HiF-1α dynamics. Then, we tested different therapeutics strategies. We show that a therapeutic decrease of HiF-1α nuclear level is not always correlated with an attenuation of reoxygenation-induced instabilities. Thus, it appears that the design of anti-HiF-1α therapies have to take into account these two aspects to maximize their efficiency.
Collapse
|
20
|
El-Chemaly S, Pacheco-Rodriguez G, Malide D, Meza-Carmen V, Kato J, Cui Y, Padilla PI, Samidurai A, Gochuico BR, Moss J. Nuclear localization of vascular endothelial growth factor-D and regulation of c-Myc-dependent transcripts in human lung fibroblasts. Am J Respir Cell Mol Biol 2014; 51:34-42. [PMID: 24450584 DOI: 10.1165/rcmb.2013-0417oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lymphangiogenesis and angiogenesis are processes that are, in part, regulated by vascular endothelial growth factor (VEGF)-D. The formation of lymphatic structures has been implicated in multiple lung diseases, including pulmonary fibrosis. VEGF-D is a secreted protein produced by fibroblasts and macrophages, which induces lymphangiogenesis by signaling via VEGF receptor-3, and angiogenesis through VEGF receptor-2. VEGF-D contains a central VEGF homology domain, which is the biologically active domain, with flanking N- and C-terminal propeptides. Full-length VEGF-D (∼ 50 kD) is proteolytically processed in the extracellular space, to generate VEGF homology domain that contains the VEGF-D receptor-binding sites. Here, we report that, independent of its cell surface receptors, full-length VEGF-D accumulated in nuclei of fibroblasts, and that this process appears to increase with cell density. In nuclei, full-length VEGF-D associated with RNA polymerase II and c-Myc. In cells depleted of VEGF-D, the transcriptionally regulated genes appear to be modulated by c-Myc. These findings have potential clinical implications, as VEGF-D was found in fibroblast nuclei in idiopathic pulmonary fibrosis, a disease characterized by fibroblast proliferation. These findings are consistent with actions of full-length VEGF-D in cellular homeostasis in health and disease, independent of its receptors.
Collapse
|
21
|
Bergeron D, Lapointe C, Bissonnette C, Tremblay G, Motard J, Roucou X. An out-of-frame overlapping reading frame in the ataxin-1 coding sequence encodes a novel ataxin-1 interacting protein. J Biol Chem 2013; 288:21824-35. [PMID: 23760502 DOI: 10.1074/jbc.m113.472654] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Spinocerebellar ataxia type 1 is an autosomal dominant cerebellar ataxia associated with the expansion of a polyglutamine tract within the ataxin-1 (ATXN1) protein. Recent studies suggest that understanding the normal function of ATXN1 in cellular processes is essential to decipher the pathogenesis mechanisms in spinocerebellar ataxia type 1. We found an alternative translation initiation ATG codon in the +3 reading frame of human ATXN1 starting 30 nucleotides downstream of the initiation codon for ATXN1 and ending at nucleotide 587. This novel overlapping open reading frame (ORF) encodes a 21-kDa polypeptide termed Alt-ATXN1 (Alternative ATXN1) with a completely different amino acid sequence from ATXN1. We introduced a hemagglutinin tag in-frame with Alt-ATXN1 in ATXN1 cDNA and showed in cell culture the co-expression of both ATXN1 and Alt-ATXN1. Remarkably, Alt-ATXN1 colocalized and interacted with ATXN1 in nuclear inclusions. In contrast, in the absence of ATXN1 expression, Alt-ATXN1 displays a homogenous nucleoplasmic distribution. Alt-ATXN1 interacts with poly(A)(+) RNA, and its nuclear localization is dependent on RNA transcription. Polyclonal antibodies raised against Alt-ATXN1 confirmed the expression of Alt-ATXN1 in human cerebellum expressing ATXN1. These results demonstrate that human ATXN1 gene is a dual coding sequence and that ATXN1 interacts with and controls the subcellular distribution of Alt-ATXN1.
Collapse
Affiliation(s)
- Danny Bergeron
- Department of Biochemistry, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1E 4K8, Canada
| | | | | | | | | | | |
Collapse
|
22
|
Zhao WT, Zhou CF, Li XB, Zhang YF, Fan L, Pelletier J, Fang J. The von Hippel-Lindau protein pVHL inhibits ribosome biogenesis and protein synthesis. J Biol Chem 2013; 288:16588-16597. [PMID: 23612971 DOI: 10.1074/jbc.m113.455121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
pVHL, the product of von Hippel-Lindau (VHL) tumor suppressor gene, functions as the substrate recognition component of an E3-ubiquitin ligase complex that targets hypoxia inducible factor α (HIF-α) for ubiquitination and degradation. Besides HIF-α, pVHL also interacts with other proteins and has multiple functions. Here, we report that pVHL inhibits ribosome biogenesis and protein synthesis. We find that pVHL associates with the 40S ribosomal protein S3 (RPS3) but does not target it for destruction. Rather, the pVHL-RPS3 association interferes with the interaction between RPS3 and RPS2. Expression of pVHL also leads to nuclear retention of pre-40S ribosomal subunits, diminishing polysomes and 18S rRNA levels. We also demonstrate that pVHL suppresses both cap-dependent and cap-independent protein synthesis. Our findings unravel a novel function of pVHL and provide insight into the regulation of ribosome biogenesis by the tumor suppressor pVHL.
Collapse
Affiliation(s)
- Wen-Ting Zhao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cheng-Fu Zhou
- Department of Surgery, Zhongshan Hospital, Fudan University School of Medicine, Shanghai 200030, China
| | - Xue-Bing Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun-Fang Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Fan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Jing Fang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
23
|
Metcalf JL, Bradshaw PS, Komosa M, Greer SN, Stephen Meyn M, Ohh M. K63-ubiquitylation of VHL by SOCS1 mediates DNA double-strand break repair. Oncogene 2013; 33:1055-65. [PMID: 23455319 DOI: 10.1038/onc.2013.22] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 12/17/2012] [Accepted: 01/11/2013] [Indexed: 01/09/2023]
Abstract
DNA repair is essential for maintaining genomic stability, and defects in this process significantly increase the risk of cancer. Clear-cell renal cell carcinoma (CCRCC) caused by inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene is characterized by high genomic instability. However, the molecular mechanism underlying the association between the loss of VHL and genomic instability remains unclear. Here, we show that suppressor of cytokine signaling 1 (SOCS1) promotes nuclear redistribution and K63-ubiquitylation of VHL in response to DNA double-strand breaks (DSBs). Loss of VHL or VHL mutations that compromise its K63-ubiquitylation attenuates the DNA-damage response (DDR), resulting in decreased homologous recombination repair and persistence of DSBs. These results identify VHL as a component of the DDR network, inactivation of which contributes to the genomic instability associated with CCRCC.
Collapse
Affiliation(s)
- J L Metcalf
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - P S Bradshaw
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - M Komosa
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - S N Greer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - M Stephen Meyn
- 1] Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada [2] Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada [3] Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - M Ohh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Lin YT, Wen WC, Yen PH. Transcription-dependent nuclear localization of DAZAP1 requires an N-terminal signal. Biochem Biophys Res Commun 2012; 428:422-6. [PMID: 23111326 DOI: 10.1016/j.bbrc.2012.10.076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 10/20/2012] [Indexed: 10/27/2022]
Abstract
Deleted in Azoospermia Associated Protein 1 (DAZAP1) is a ubiquitous hnRNP protein required for normal development and spermatogenesis. It resides predominantly in the nucleus and moves between the nucleus and the cytoplasm via a ZNS shuttling signal at its C-terminus. DAZAP1 accumulates in the cytoplasm when RNA polymerase II activity is inhibited by actinomycin D. Here we report the mapping of a 42-amino acid segment (N42) at the N-terminus of DAZAP1 that is both necessary and sufficient for its transcription-dependent nuclear localization. In addition, using a yeast two-hybrid system, we have identified SLIRP as a N42-binding protein which may regulate DAZAP1 subcellular localization.
Collapse
Affiliation(s)
- Yi-Tzu Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | | | | |
Collapse
|
25
|
|
26
|
Abstract
Ninety percent or more of kidney cancers are believed to be of epithelial cell origin, and are referred to as renal cell carcinoma (RCC), which are further subdivided based on histology into clear-cell RCC (75%), papillary RCC (15%), chromophobe tumor (5%), and oncocytoma (5%). Some genes confer an increased risk of these various histologic RCC subtypes. In practice, there is some overlap among the histologic subtypes, and there are some shared molecular features among these tumor types. This review focuses primarily on the most common form of RCC, clear-cell renal carcinoma, noting some recent advances in the other histologic subtypes.
Collapse
Affiliation(s)
- Lianjie Li
- Howard Hughes Medical Institute (HHMI), 4000 Jones Bridge Road, Chevy Chase, MD 20815, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, 450 Brookline Avenue, Boston, MA 02215, USA
| | - William G. Kaelin
- Howard Hughes Medical Institute (HHMI), 4000 Jones Bridge Road, Chevy Chase, MD 20815, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
27
|
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: 0.1] [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
|
28
|
Li M, Kim WY. Two sides to every story: the HIF-dependent and HIF-independent functions of pVHL. J Cell Mol Med 2011; 15:187-95. [PMID: 21155973 PMCID: PMC3229769 DOI: 10.1111/j.1582-4934.2010.01238.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 10/04/2010] [Indexed: 01/15/2023] Open
Abstract
von Hippel-Lindau (VHL) disease is a hereditary cancer syndrome caused by inherited mutations that inactivate the VHL tumour suppressor gene. The VHL locus encodes pVHL, whose best studied function is to bind to and down-regulate the hypoxia-inducible factor (HIF) family of oxygen-dependent transcription factors. Early efforts have established the fundamental role of HIF in VHL-defective tumorigenesis and in particular renal cell carcinoma. However, recent findings have revealed an alternate side to the story, the HIF-independent tumour suppressor functions of pVHL. These include pVHL's ability to regulate apoptosis and senescence as well as its role in the maintenance of primary cilium and orchestrating the deposition of the extracellular matrix. To what extent these HIF-dependent and HIF-independent functions cooperate in VHL-defective tumorigenesis remains to be determined.
Collapse
Affiliation(s)
- Mingqing Li
- Departments of Medicine and Genetics, Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - William Y Kim
- Departments of Medicine and Genetics, Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC, USA
| |
Collapse
|
29
|
Cai Q, Robertson ES. Ubiquitin/SUMO modification regulates VHL protein stability and nucleocytoplasmic localization. PLoS One 2010; 5. [PMID: 20844582 PMCID: PMC2936558 DOI: 10.1371/journal.pone.0012636] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 08/14/2010] [Indexed: 01/08/2023] Open
Abstract
Functional inactivation of the von Hippel-Lindau (VHL) tumor suppressor protein is linked to the development of several forms of cancer as well as oncogenic progression like sporadic renal clear-cell carcinomas (RCC). Despite the critical role played by VHL in destruction of hypoxia-inducible factor α (HIFα) via ubiquitin-mediated proteolysis, very little is known about the post-translational modification which regulates VHL activity. Our previous study showed that the SUMO E3 ligase PIASy interacts with VHL and induces VHL SUMOylation on lysine residue 171 (Cai et al, PLoS ONE, 2010, 5(3):e9720). Here we further report that VHL also undergoes ubiquitylation on both lysine residues 171 and 196, which is blocked by PIASy. Moreover, using a VHL-SUMO1 or ubiquitin fusion protein, we found that ubiquitylated VHL is localized predominantly in the cytoplasm, while SUMOylated VHL results in increased VHL protein stability and nuclear redistribution. Interestingly, substitution of lysine 171 and 196 to arginine of VHL abrogates its inhibitory function on the transcriptional activity of HIFα, and tube formation in vitro. This demonstrates that post-translational modifications like ubiquitylation and SUMOylation contributes to VHL protein stability and nucleocytoplasmic shuttling, and that the overall function of VHL in tumor suppression may require a precise and dynamically regulated process which involves protein modification.
Collapse
Affiliation(s)
- Qiliang Cai
- Department of Microbiology and Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Erle S. Robertson
- Department of Microbiology and Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
30
|
Kim JJ, Rini BI, Hansel DE. Von Hippel Lindau syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 685:228-49. [PMID: 20687511 DOI: 10.1007/978-1-4419-6448-9_22] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Von Hippel-Lindau syndrome (VHLS) is an autosomal dominant familial cancer syndrome arising from germ-line inactivation of the VHL gene on the short arm of chromosome 3. VHLS manifests in a myriad of hyper-vascular tumors of both benign and malignant nature. Incidence of VHLS is roughly 1 in 36,000 live births and has over 90% penetrance by the age of 65. Improved understanding of the natural history and biology of VHLS has led to the introduction of screening protocols, early interventions and improved treatments, all of which resulted in a substantially improved prognosis for this disease. Further details regardingvariegated molecular pathways and mechanisms ofVHLS are emerging with the subsequent advent of novel treatment protocols that are currently in clinical trials.
Collapse
Affiliation(s)
- Jenny J Kim
- Cleveland Clinic, Taussig Cancer Institute, Celveland, Ohio 44195, USA
| | | | | |
Collapse
|
31
|
Boateng SY, Senyo SE, Qi L, Goldspink PH, Russell B. Myocyte remodeling in response to hypertrophic stimuli requires nucleocytoplasmic shuttling of muscle LIM protein. J Mol Cell Cardiol 2009; 47:426-35. [PMID: 19376126 PMCID: PMC2739242 DOI: 10.1016/j.yjmcc.2009.04.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 03/19/2009] [Accepted: 04/07/2009] [Indexed: 12/14/2022]
Abstract
CSRP3 or muscle LIM protein (MLP) is a nucleocytoplasmic shuttling protein and a mechanosensor in cardiac myocytes. MLP regulation and function was studied in cultured neonatal rat myocytes treated with pharmacological or mechanical stimuli. Either verapamil or BDM decreased nuclear MLP while phenylephrine and cyclic strain increased it. These results suggest that myocyte contractility regulates MLP subcellular localization. When RNA polymerase II was inhibited with alpha-amanitin, nuclear MLP was reduced by 30%. However, when both RNA polymerase I and II were inhibited with actinomycin D, there was a 90% decrease in nuclear MLP suggesting that its nuclear translocation is regulated by both nuclear and nucleolar transcriptional activity. Using cell permeable synthetic peptides containing the putative nuclear localization signal (NLS) of MLP, nuclear import of the protein in cultured rat neonatal myocytes was inhibited. The NLS of MLP also localizes to the nucleolus. Inhibition of nuclear translocation prevented the increased protein accumulation in response to phenylephrine. Furthermore, cyclic strain of myocytes after prior NLS treatment to remove nuclear MLP resulted in disarrayed sarcomeres. Increased protein synthesis and brain natriuretic peptide expression were also prevented suggesting that MLP is required for remodeling of the myofilaments and gene expression. These findings suggest that nucleocytoplasmic shuttling MLP plays an important role in the regulation of the myocyte remodeling and hypertrophy and is required for adaptation to hypertrophic stimuli.
Collapse
Affiliation(s)
- Samuel Y Boateng
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago IL 60612-7342. USA
- Institute for Cardiovascular and Metabolic Research, Schools of Biological Sciences and Pharmacy, University of Reading, Reading, Berkshire RG66UB, United Kingdom
| | - Samuel E Senyo
- Department of Bioengineering at University of Illinois at Chicago, 851 S Morgan St Chicago, IL 60607-7052
| | - Lixin Qi
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago IL 60612-7342. USA
| | - Paul H Goldspink
- Section of Cardiology, University of Illinois at Chicago, 840 South Wood St, Chicago IL 60612-7342
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago IL 60612-7342. USA
| |
Collapse
|
32
|
Real-time imaging of HIF-1alpha stabilization and degradation. PLoS One 2009; 4:e5077. [PMID: 19347037 PMCID: PMC2660410 DOI: 10.1371/journal.pone.0005077] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 02/25/2009] [Indexed: 01/09/2023] Open
Abstract
HIF-1α is overexpressed in many human cancers compared to normal tissues due to the interaction of a multiplicity of factors and pathways that reflect specific genetic alterations and extracellular stimuli. We developed two HIF-1α chimeric reporter systems, HIF-1α/FLuc and HIF-1α(ΔODDD)/FLuc, to investigate the tightly controlled level of HIF-1α protein in normal (NIH3T3 and HEK293) and glioma (U87) cells. These reporter systems provided an opportunity to investigate the degradation of HIF-1α in different cell lines, both in culture and in xenografts. Using immunofluorescence microscopy, we observed different patterns of subcellular localization of HIF-1α/FLuc fusion protein between normal cells and cancer cells; similar differences were observed for HIF-1α in non-transduced, wild-type cells. A dynamic cytoplasmic-nuclear exchange of the fusion protein and HIF-1α was observed in NIH3T3 and HEK293 cells under different conditions (normoxia, CoCl2 treatment and hypoxia). In contrast, U87 cells showed a more persistent nuclear localization pattern that was less affected by different growing conditions. Employing a kinetic model for protein degradation, we were able to distinguish two components of HIF-1α/FLuc protein degradation and quantify the half-life of HIF-1α fusion proteins. The rapid clearance component (t1/2 ∼4–6 min) was abolished by the hypoxia-mimetic CoCl2, MG132 treatment and deletion of ODD domain, and reflects the oxygen/VHL-dependent degradation pathway. The slow clearance component (t1/2 ∼200 min) is consistent with other unidentified non-oxygen/VHL-dependent degradation pathways. Overall, the continuous bioluminescence readout of HIF-1α/FLuc stabilization in vitro and in vivo will facilitate the development and validation of therapeutics that affect the stability and accumulation of HIF-1α.
Collapse
|
33
|
Khacho M, Lee S. Subcellular dynamics of the VHL tumor suppressor: on the move for HIF degradation. Future Oncol 2009; 5:85-95. [DOI: 10.2217/14796694.5.1.85] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The von Hippel–Lindau (VHL) tumor suppressor protein, the recognition component of an E3 ubiquitin ligase complex, recruits the α-subunit of the hypoxia-inducible factor (HIFα) for oxygen-dependent degradation. The ability of VHL to mediate efficient degradation of HIFα is also dependent on its oxygen/pH-regulated subcellular trafficking. Under aerobic conditions, VHL engages in nuclear–cytoplasmic trafficking that requires ongoing transcription and is mediated by a novel nuclear export motif, the transcription-dependent nuclear export motif (TD-NEM). Disease-causing mutations targeting TD-NEM restrain VHL from mediating efficient oxygen-dependent degradation of HIFα by altering its subcellular dynamics. In addition, decreasing the extracellular pH, during anaerobic metabolism, stabilizes HIFα by triggering the relocalization and static detention of VHL to nucleoli. Together, these recent findings support the critical role of subcellular trafficking and dynamic properties for the function of VHL in promoting HIF regulation and tumor suppression.
Collapse
Affiliation(s)
- Mireille Khacho
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Stephen Lee
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
34
|
Spielberger JC, Moody AD, Watson WH. Oxidation and nuclear localization of thioredoxin-1 in sparse cell cultures. J Cell Biochem 2008; 104:1879-89. [PMID: 18384140 DOI: 10.1002/jcb.21762] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Reactive oxygen species (ROS) were once viewed only as mediators of toxicity, but it is now recognized that they also contribute to redox signaling through oxidation of specific cysteine thiols on regulatory proteins. Cells in sparse cultures have increased ROS relative to confluent cultures, but it is not known whether protein redox states are affected under these conditions. The purpose of the present study was to determine whether culture conditions affect the redox state of thioredoxin-1 (Trx1), the protein responsible for reducing most oxidized proteins in the cytoplasm and nucleus. The results showed that Trx1 was more oxidized in sparse HeLa cell cultures than in confluent cells. The glutathione pool was also more oxidized, demonstrating that both of the major cellular redox regulating systems were affected by culture density. In addition, the total amount of Trx1 protein was lower and the subcellular distribution of Trx1 was different in sparse cells. Trx1 in sparse cultures was predominantly nuclear whereas it was predominantly cytoplasmic in confluent cultures. This localization pattern was not unique to HeLa cells as it was also observed in A549, Cos-1 and HEK293 cells. These findings demonstrate that Trx1 is subject to changes in expression, redox state and subcellular localization with changing culture density, indicating that the redox environments of the cytoplasm and the nucleus are distinct and have different requirements under different culture conditions.
Collapse
Affiliation(s)
- Jeanine C Spielberger
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
35
|
Khacho M, Mekhail K, Pilon-Larose K, Pause A, Côté J, Lee S. eEF1A is a novel component of the mammalian nuclear protein export machinery. Mol Biol Cell 2008; 19:5296-308. [PMID: 18799616 DOI: 10.1091/mbc.e08-06-0562] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The cytoplasmic translation factor eEF1A has been implicated in the nuclear export of tRNA species in lower eukaryotes. Here we demonstrate that eEF1A plays a central role in nuclear export of proteins in mammalian cells. TD-NEM (transcription-dependent nuclear export motif), a newly characterized nuclear export signal, mediates efficient nuclear export of several proteins including the von Hippel-Lindau (VHL) tumor suppressor and the poly(A)-binding protein (PABP1) in a manner that is dependent on ongoing RNA polymerase II (RNA PolII)-dependent transcription. eEF1A interacts specifically with TD-NEM of VHL and PABP1 and disrupting this interaction, by point mutations of key TD-NEM residues or treatment with actinomycin D, an inhibitor of RNA PolII-dependent transcription, prevents assembly and nuclear export. siRNA-induced knockdown or antibody-mediated depletion of eEF1A prevents in vivo and in vitro nuclear export of TD-NEM-containing proteins. Nuclear retention experiments and inhibition of the Exportin-5 pathway suggest that eEF1A stimulates nuclear export of proteins from the cytoplasmic side of the nuclear envelope, without entering the nucleus. Together, these data identify a role for eEF1A, a cytoplasmic mediator of tRNA export in yeast, in the nuclear export of proteins in mammalian cells. These results also provide a link between the translational apparatus and subcellular trafficking machinery demonstrating that these two central pathways in basic metabolism can act cooperatively.
Collapse
Affiliation(s)
- Mireille Khacho
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | | | | | | | | | | |
Collapse
|
36
|
Manuel Trigo J, Bellmunt J. Estrategias actuales en el tratamiento del carcinoma de células renales: fármacos dirigidos a dianas moleculares. Med Clin (Barc) 2008; 130:380-92. [DOI: 10.1157/13117476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
37
|
Nuclear expression of hypoxia-inducible factor-1alpha in clear cell renal cell carcinoma is involved in tumor progression. Am J Surg Pathol 2008; 31:1875-81. [PMID: 18043042 DOI: 10.1097/pas.0b013e318094fed8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The most frequent genomic abnormality in clear cell renal cell carcinoma (cc-RCC) is inactivation of Von Hippel-Lindau gene (VHL). pVHL19 is a ligase promoting proteosomal degradation of hypoxia-inducible factor-1alpha (HIF-1alpha); pVHL30 is associated with microtubules. VHL exert its oncogenetic action both directly and through HIF-1alpha activation. TNM classification is unable to define a correct prognostic evaluation of intracapsular cc-RCC. The nucleo-cytoplasmic trafficking in VHL/HIF-1alpha pathway could be relevant in understanding the molecular pathogenesis of renal carcinogenesis. This study analyzes VHL/HIF-1alpha proteins in a large series of intracapsular cc-RCCs, correlating their expression and cellular localization with prognosis. MATERIALS AND METHODS Two anti-pVHL (clones Ig32 and Ig33) and 1 anti-HIF-1alpha were used on tissue microarrays from 136 intracapsular cc-RCCs (mean follow-up: 74 mo). Clone 32 recognizes both pVHLs, whereas clone 33 only pVHL30. Results were matched with clinicopathologic variables and tumor-specific survival (TSS). RESULTS A strong cytoplasmic positivity was found for all antibodies in the largest part of cases, associated to a strong nuclear localization in the case of HIF-1alpha. All pVHL-negative cases were associated with high HIF-1alpha expression. pVHL negativity and HIF-1alpha nuclear positivity significantly correlated with shorter TSS. In multivariate analysis both pVHL negativity and HIF-1alpha nuclear expression were independent predictors of TSS. CONCLUSIONS The localization of the proteins well matches with their role and with the supposed tumor molecular pathways. The correlation with prognosis of VHL/HIF-1alpha alterations confirms the relevance of their molecular pathway and of the cellular trafficking of their products in the pathogenesis of renal cancer.
Collapse
|
38
|
Mobility of the von Hippel-Lindau tumour suppressor protein is regulated by kinesin-2. Exp Cell Res 2008; 314:1229-36. [PMID: 18261724 DOI: 10.1016/j.yexcr.2007.12.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 11/23/2007] [Accepted: 12/20/2007] [Indexed: 12/31/2022]
Abstract
The von Hippel-Lindau tumour suppressor protein (pVHL) participates in many cellular processes including oxygen sensing, microtubule stability and primary cilia regulation. Recently, we identified ATP-dependent motor complex kinesin-2 to endogenously bind the full-length variant of VHL (pVHL30) in primary kidney cells, and mediate its association to microtubules. Here we show that pVHL also endogenously binds the neuronal kinesin-2 complex, which slightly differs from renal kinesin-2. To investigate the role of kinesin-2 in pVHL mobility, we performed fluorescence recovery after photobleaching (FRAP) experiments in neuroblastoma cells. We observe that pVHL30 is a highly mobile cytoplasmic protein, which becomes an immobile centrosomal protein after ATP-depletion in living cells. This response to ATP-depletion is independent of GSK3beta-dependent phosphorylation of pVHL30. Furthermore, VHL variant alleles with reduced binding to kinesin-2 fail to respond to ATP-depletion. Accordingly, interfering with pVHL30-KIF3A interaction by either overexpressing a dominant negative construct or by reducing endogenous cellular levels of KIF3A by RNAi abolishes pVHL's response to ATP-depletion. From these data we suggest that mobility of a subcellular pool of pVHL is regulated by the ATP-dependent kinesin-2 motor. Kinesin-2 driven mobility of cytoplasmic pVHL might enable pVHL to function as a tumour suppressor.
Collapse
|
39
|
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: 0.7] [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
|
40
|
Cinelli RAG, Ferrari A, Pellegrini V, Tyagi M, Giacca M, Beltram F. The Enhanced Green Fluorescent Protein as a Tool for the Analysis of Protein Dynamics and Localization: Local Fluorescence Study at the Single-molecule Level. Photochem Photobiol 2007. [DOI: 10.1562/0031-8655(2000)0710771tegfpa2.0.co2] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
41
|
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
|
42
|
Mylonis I, Chachami G, Samiotaki M, Panayotou G, Paraskeva E, Kalousi A, Georgatsou E, Bonanou S, Simos G. Identification of MAPK phosphorylation sites and their role in the localization and activity of hypoxia-inducible factor-1alpha. J Biol Chem 2006; 281:33095-106. [PMID: 16954218 DOI: 10.1074/jbc.m605058200] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1) controls the expression of most genes induced by hypoxic conditions. Regulation of expression and activity of its inducible subunit, HIF-1alpha, involves several post-translational modifications. To study HIF-1alpha phosphorylation, we have used human full-length recombinant HIF-1alpha as a substrate in kinase assays. We show that at least two different nuclear protein kinases, one of them identified as p42/p44 MAPK, can modify HIF-1alpha. Analysis of in vitro phosphorylated HIF-1alpha by mass spectroscopy revealed residues Ser-641 and Ser-643 as possible MAPK phosphorylation sites. Site-directed mutagenesis of these residues reduced significantly the phosphorylation of HIF-1alpha. When these mutant forms of HIF-1alpha were expressed in HeLa cells, they exhibited much lower transcriptional activity than the wild-type form. However, expression of the same mutants in yeast revealed that their capacity to stimulate transcription was not significantly compromised. Localization of the green fluorescent protein-tagged HIF-1alpha mutants in HeLa cells showed their exclusion from the nucleus in contrast to wild-type HIF-1alpha. Treatment of the cells with leptomycin B, an inhibitor of the major exportin CRM1, reversed this exclusion and led to nuclear accumulation and partial recovery of the activity of the HIF-1alpha mutants. Moreover, inhibition of the MAPK pathway by PD98059 impaired the phosphorylation, nuclear accumulation, and activity of wild-type GFP-HIF-1alpha. Overall, these data suggest that phosphorylation of Ser-641/643 by MAPK promotes the nuclear accumulation and transcriptional activity of HIF-1alpha by blocking its CRM1-dependent nuclear export.
Collapse
Affiliation(s)
- Ilias Mylonis
- Laboratory of Biochemistry, Department of Medicine, University of Thessaly, Larissa 41222, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yoshikawa Y, Morimatsu M, Ochiai K, Nagano M, Yamane Y, Tomizawa N, Sasaki N, Hashizume K. Insertion/deletion polymorphism in the BRCA2 nuclear localization signal. Biomed Res 2006; 26:109-16. [PMID: 16011303 DOI: 10.2220/biomedres.26.109] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Mutations in human BRCA2 confer an increased risk of female breast cancer. In this study, we found a novel insertion/deletion polymorphism (10204insAAA causing amino acid change M3332IK) in canine BRCA2, which is located in the putative second nuclear localization signal (NLS2) and C-terminal Rad51-binding region. The nuclear localization of the insAAA C-terminus was more efficient than localization of the delAAA sequence when NLS1 was mutated. Strong, comparable Rad51 binding was observed for both the insAAA and delAAA C-termini. Dogs with the insertion/deletion polymorphism will provide a new model for studying the function of BRCA2.
Collapse
Affiliation(s)
- Yasunaga Yoshikawa
- Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka 020-8550, Japan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Sun X, Liu M, Wei Y, Liu F, Zhi X, Xu R, Krissansen GW. Overexpression of von Hippel-Lindau tumor suppressor protein and antisense HIF-1alpha eradicates gliomas. Cancer Gene Ther 2006; 13:428-435. [PMID: 16211089 DOI: 10.1038/sj.cgt.7700907] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Revised: 08/08/2005] [Accepted: 08/12/2005] [Indexed: 11/08/2022]
Abstract
The von Hippel-Lindau tumor suppressor protein (pVHL) suppresses tumor formation by binding the alpha subunits of hypoxia-inducible-factors responsible for stimulating tumor angiogenesis and glycolysis, and targeting them for ubiquitination and proteasomal destruction. Loss of pVHL leads to tumorigenesis and development of sporadic renal cell carcinomas and central nervous system hemangioblastomas. In the present study, we investigated whether engineered overexpression of pVHL in C6 glioma cells, which already express endogenous pVHL, would suppress the tumorigenicity of this particular tumor cell type. C6 cells overexpressing VHL displayed a reduced growth rate (70% inhibition) compared to the parental cell line when subcutaneously implanted in athymic (nu/nu) mice. Growth inhibition was associated with a 50% reduction in the number of tumor vessels and a 60% increase in tumor cell apoptosis, due in part to downregulation of HIF-1, VEGF, and the antiapoptotic factor Bcl-2, respectively. Gene transfer of VHL suppressed the growth of established C6 gliomas, and synergized with antisense HIF-1 to completely eradicate tumors. The data suggest that VHL gene therapy and/or agents that increase VHL expression could have utility in the treatment of gliomas, particularly when combined with agents that inhibit the expression or function of HIF-1.
Collapse
Affiliation(s)
- X Sun
- Department of Molecular Medicine and Pathology, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
45
|
Kumar S, Saradhi M, Chaturvedi NK, Tyagi RK. Intracellular localization and nucleocytoplasmic trafficking of steroid receptors: an overview. Mol Cell Endocrinol 2006; 246:147-56. [PMID: 16388893 DOI: 10.1016/j.mce.2005.11.028] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Subcellular compartmentalization and dynamic movements of steroid receptors are major steps in executing their transcription regulatory function. Though significant progress has been made in understanding the mechanisms underlying nuclear import of NLS-bearing proteins, our general and mechanistic understanding about the nuclear export processes has begun to emerge only recently. The discovery of most commonly utilized CRM1/exportin1 dependent nuclear export pathway is attributed to a potent nuclear export inhibitor leptomycin B that helped dissecting this and other nuclear export pathways. Simultaneously, utilization of green fluorescent protein (GFP)-tagged intracellular steroid receptors has contributed to not only resolving controversial issue of subcellular localization of unliganded hormone receptors but also provided further insight into finer details of receptor dynamics in living cells. With judicious use of leptomycin B and expression of GFP-tagged receptors in living cells, existence of exportin1/CRM1 independent pathway(s), nuclear export signals and receptors for bi-directional translocation that are unique to steroid receptor trafficking have been specified. Currently, we appear to be arriving at a consensus that steroid/nuclear receptors follow dynamic nucleocytoplasmic processes that deviate from the ones commonly utilized by majority of other proteins.
Collapse
Affiliation(s)
- Sanjay Kumar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | | | | | | |
Collapse
|
46
|
O'Hare MJ, Kushwaha N, Zhang Y, Aleyasin H, Callaghan SM, Slack RS, Albert PR, Vincent I, Park DS. Differential roles of nuclear and cytoplasmic cyclin-dependent kinase 5 in apoptotic and excitotoxic neuronal death. J Neurosci 2006; 25:8954-66. [PMID: 16192386 PMCID: PMC6725602 DOI: 10.1523/jneurosci.2899-05.2005] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cyclin-dependent kinase 5 (cdk5) is a member of the cyclin-dependent kinase family whose activity is localized mainly to postmitotic neurons attributable to the selective expression of its activating partners p35 and p39. Deregulation of cdk5, as a result of calpain cleavage of p35 to a smaller p25 form, has been suggested to be a central component of neuronal death underlying numerous neurodegenerative diseases. However, the relevance of cdk5 in apoptotic death that relies on the mitochondrial pathway is unknown. Furthermore, evidence that cdk5 can also promote neuronal survival has necessitated a more complex understanding of cdk5 in the control of neuronal fate. Here we explore each of these issues using apoptotic and excitotoxic death models. We find that apoptotic death induced by the DNA-damaging agent camptothecin is associated with early transcription-mediated loss of p35 and with late production of p25 that is dependent on Bax, Apaf1, and caspases. In contrast, during excitotoxic death induced by glutamate, neurons rapidly produce p25 independent of the mitochondrial pathway. Analysis of the localization of p35 and p25 revealed that p35 is mainly cytoplasmic, whereas p25 accumulates selectively in the nucleus. By targeting a dominant-negative cdk5 to either the cytoplasm or nucleus, we show that cdk5 has a death-promoting activity within the nucleus and that this activity is required in excitotoxic death but not apoptotic death. Moreover, we also find that cdk5 contributes to pro-survival signaling selectively within the cytoplasm, and manipulation of this signal can modify death induced by both excitotoxicity and DNA damage.
Collapse
Affiliation(s)
- Michael J O'Hare
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa Health Research Institute, Ottawa, Ontario, K1H 8M5, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mekhail K, Khacho M, Carrigan A, Hache RRJ, Gunaratnam L, Lee S. Regulation of ubiquitin ligase dynamics by the nucleolus. ACTA ACUST UNITED AC 2005; 170:733-44. [PMID: 16129783 PMCID: PMC2171338 DOI: 10.1083/jcb.200506030] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Cellular pathways relay information through dynamic protein interactions. We have assessed the kinetic properties of the murine double minute protein (MDM2) and von Hippel-Lindau (VHL) ubiquitin ligases in living cells under physiological conditions that alter the stability of their respective p53 and hypoxia-inducible factor substrates. Photobleaching experiments reveal that MDM2 and VHL are highly mobile proteins in settings where their substrates are efficiently degraded. The nucleolar architecture converts MDM2 and VHL to a static state in response to regulatory cues that are associated with substrate stability. After signal termination, the nucleolus is able to rapidly release these proteins from static detention, thereby restoring their high mobility profiles. A protein surface region of VHL's β-sheet domain was identified as a discrete [H+]-responsive nucleolar detention signal that targets the VHL/Cullin-2 ubiquitin ligase complex to nucleoli in response to physiological fluctuations in environmental pH. Data shown here provide the first evidence that cells have evolved a mechanism to regulate molecular networks by reversibly switching proteins between a mobile and static state.
Collapse
Affiliation(s)
- Karim Mekhail
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Derheimer FA, Chang CW, Ljungman M. Transcription inhibition: a potential strategy for cancer therapeutics. Eur J Cancer 2005; 41:2569-76. [PMID: 16213135 DOI: 10.1016/j.ejca.2005.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Interference with transcription triggers a stress response leading to the induction of the tumour suppressor p53. If transcription is not restored within a certain time frame cells may undergo apoptosis in a p53-dependent and independent manner. The mechanisms by which blockage of transcription induces apoptosis may involve diminished levels of anti-apoptotic factors, inappropriate accumulation of proteins in the nucleus, accumulation of p53 at mitochondria or complications during replication. Many chemotherapeutic agents currently used in the clinic interfere with transcription and this interference may contribute to their anti-cancer activities. Future efforts should be directed towards exploring whether interference of transcription could be used as an anti-cancer therapeutic strategy.
Collapse
Affiliation(s)
- Frederick A Derheimer
- Department of Radiation Oncology, Division of Radiation and Cancer Biology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
49
|
Paltoglou SM, Roberts BJ. Role of the von Hippel-Lindau tumour suppressor protein in the regulation of HIF-1alpha and its oxygen-regulated transactivation domains at high cell density. Oncogene 2005; 24:3830-5. [PMID: 15750626 DOI: 10.1038/sj.onc.1208531] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hypoxia-inducible factor-1alpha (HIF-1alpha) induction and associated transcription were investigated during high cell density, focusing on the negative regulator of HIF-1alpha expression, the von Hippel-Lindau (VHL) protein. In 293T and HeLa cells, HIF-1alpha protein levels and associated transcription were induced as cells approached confluence. To determine whether these changes were due to a deficit in nuclear VHL-mediated ubiquitination of HIF-1alpha at confluence, cells were stably transfected with VHL. Overexpression of VHL in 293T cells had no demonstrable effect on the induction and nuclear accumulation of HIF-1alpha during high cell density or associated transcription. Moreover, RCC cells stably transfected with full-length VHL failed to exhibit the cell-density-dependent induction of HIF-1alpha noted in other cell lines. Investigation of both N-terminal and C-terminal (aa 727-826) oxygen-regulated proline and asparagine hydroxylation of HIF-1alpha revealed that both are inhibited during high cell density, as determined by impaired capture of HIF-1alpha by VHL and enhanced C-terminal transactivation. Finally, cell-density-mediated induction of HIF-1alpha and GLUT1 in RCC cells could be completely reconstituted by mutations in VHL binding affinity, suggesting that cell-density dependent induction of HIF-1alpha and transactivation may underpin some of the deregulated gene expression observed in VHL disease.
Collapse
Affiliation(s)
- Steve M Paltoglou
- School of Pharmaceutical, Molecular and Biomedical Sciences, University of South Australia, Reid Building, Frome Rd., Adelaide 5000, Australia
| | | |
Collapse
|
50
|
Abstract
Germline inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene causes the von Hippel-Lindau hereditary cancer syndrome, and somatic mutations of this gene have been linked to the development of sporadic hemangioblastomas and clear-cell renal carcinomas. The VHL tumor suppressor protein (pVHL), through its oxygen-dependent polyubiquitylation of hypoxia-inducible factor (HIF), plays a central role in the mammalian oxygen-sensing pathway. This interaction between pVHL and HIF is governed by post-translational prolyl hydroxylation of HIF in the presence of oxygen by a conserved family of Egl-nine (EGLN) enzymes. In the absence of pVHL, HIF becomes stabilized and is free to induce the expression of its target genes, many of which are important in regulating angiogenesis, cell growth, or cell survival. Moreover, preliminary data indicate that HIF plays a critical role in pVHL-defective tumor formation, raising the possibility that drugs directed against HIF or its downstream targets (such as vascular endothelial growth factor) might one day play a role in the treatment of hemangioblastoma and renal cell carcinoma. On the other hand, clear genotype-phenotype correlations are emerging in VHL disease and can be rationalized if pVHL has functions separate from its control of HIF.
Collapse
Affiliation(s)
- William Y Kim
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|