1
|
Sun J, Xie L, Lv J, Zhang W, Lv J, Liang Y, Geng Y, Li X. Inhibitor of growth 4 inhibits cell proliferation, migration, and induces apoptosis of renal cell carcinoma cells. J Cell Biochem 2018; 120:6709-6717. [PMID: 30390334 DOI: 10.1002/jcb.27967] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Jiping Sun
- Department of Nephrology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| | - Liyi Xie
- Department of Nephrology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| | - Jing Lv
- Department of Nephrology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| | - Wenjing Zhang
- Department of Nephrology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| | - Jia Lv
- Department of Nephrology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| | - Yu Liang
- Department of Nephrology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| | - Yingzhou Geng
- Department of Nephrology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| | - Xudong Li
- Department of Urology The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an China
| |
Collapse
|
2
|
Han J, Lachance C, Ricketts MD, McCullough CE, Gerace M, Black BE, Côté J, Marmorstein R. The scaffolding protein JADE1 physically links the acetyltransferase subunit HBO1 with its histone H3-H4 substrate. J Biol Chem 2018; 293:4498-4509. [PMID: 29382722 DOI: 10.1074/jbc.ra117.000677] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/23/2018] [Indexed: 12/31/2022] Open
Abstract
The human enzyme histone acetyltransferase binding to ORC1 (HBO1) regulates DNA replication, cell proliferation, and development. HBO1 is part of a multiprotein histone acetyltransferase (HAT) complex that also contains inhibitor of growth family member (ING) 4/5, MYST/Esa1-associated factor (MEAF) 6, and the scaffolding proteins Jade family PHD finger (JADE) 1/2/3 or bromodomain and PHD finger-containing protein (BRPF) 2/3 to acetylate histone H4 H4K5/8/12 or H3K14, respectively. Within this four-protein complex, JADE1 determines histone H4 substrate specificity of the HBO1-HAT complex. However, the mechanism by which JADE1 controls the H4-specific acetyltransferase activity of HBO1 is unknown. Here we used recombinant proteins in vitro to dissect the specific regions and activities of HBO1 and JADE1 that mediate histone H3-H4 acetylation via the HBO1-HAT domain. We found that JADE1 increases the catalytic efficiency of HBO1 acetylation of an H3-H4 substrate by about 5-fold through an N-terminal, 21-residue HBO1- and histone-binding domain and a nearby second histone core-binding domain. We also demonstrate that HBO1 contains an N-terminal histone-binding domain (HBD) that makes additional contacts with H3-H4 independent of JADE1 interactions with histones and that the HBO1 HBD does not significantly contribute to HBO1's overall HAT activity. Experiments with JADE1 deletions in vivo recapitulated these in vitro interactions and their roles in HBO1 histone acetylation activity. Together, these results indicate that the N-terminal region of JADE1 functions as a platform that brings together the catalytic HBO1 subunit with its cognate H3-H4 substrate for histone acetylation.
Collapse
Affiliation(s)
- Joseph Han
- From the Department of Biochemistry and Biophysics.,Abramson Family Cancer Research Institute, and.,Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| | - Catherine Lachance
- the Laval University Cancer Research Center, CHU de Québec Research Center-Oncology Axis, Quebec City, Quebec G1R 3S3, Canada
| | - M Daniel Ricketts
- From the Department of Biochemistry and Biophysics.,Abramson Family Cancer Research Institute, and.,Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, and
| | - Cheryl E McCullough
- From the Department of Biochemistry and Biophysics.,Abramson Family Cancer Research Institute, and.,Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| | | | - Ben E Black
- From the Department of Biochemistry and Biophysics
| | - Jacques Côté
- the Laval University Cancer Research Center, CHU de Québec Research Center-Oncology Axis, Quebec City, Quebec G1R 3S3, Canada
| | - Ronen Marmorstein
- From the Department of Biochemistry and Biophysics, .,Abramson Family Cancer Research Institute, and.,Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| |
Collapse
|
3
|
Wang Y, Wang T, Han Y, Wu H, Zhao W, Tong D, Wei L, Zhong Z, An R, Wang Y. Reduced ING4 Expression Is Associated with the Malignancy of Human Bladder. Urol Int 2015; 94:464-71. [DOI: 10.1159/000364832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/25/2014] [Indexed: 11/19/2022]
Abstract
Introduction: Inhibitor of growth 4 (ING4) is a tumor suppressor. However the role of ING4 in human bladder malignancy is unknown. In this study, ING4 expression in human bladder cancer and its potential effects were studied. Materials and Methods: ING4 expression in 47 human bladder cancer tissues and paired adjacent normal tissues was detected by Western blotting, quantitative reverse transcription-polymerase chain reaction, and immunohistochemistry. The migration and cell cycle progression of SV-HUC-1 and T24 cells with aberrant ING4 expression were examined. Results: ING4 protein and mRNA were significantly decreased in bladder cancer tissues. ING4 protein level was significantly lower in the group of patients over 50 years of age. ING4 knockdown caused more rapid cell migration and increased the population of SV-HUC-1 and T24 cells in the G2-M phase. Conclusion: Our data suggest a close connection between aberrant ING4 expression and the carcinogenesis of human bladder cells. ING4 may be a potential target for bladder cancer chemotherapy.
Collapse
|
4
|
Klein BJ, Lalonde ME, Côté J, Yang XJ, Kutateladze TG. Crosstalk between epigenetic readers regulates the MOZ/MORF HAT complexes. Epigenetics 2013; 9:186-93. [PMID: 24169304 DOI: 10.4161/epi.26792] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The MOZ/MORF complexes represent an example of a chromatin-binding assembly whose recruitment to specific genomic regions and activity can be fine-tuned by posttranslational modifications of histones. Here we detail the structures and biological functions of epigenetic readers present in the four core subunits of the MOZ/MORF complexes, highlight the imperative role of combinatorial readout by the multiple readers, and discuss new research directions to advance our understanding of histone acetylation.
Collapse
Affiliation(s)
- Brianna J Klein
- Department of Pharmacology; University of Colorado School of Medicine; Aurora, CO USA
| | - Marie-Eve Lalonde
- Laval University Cancer Research Center; Hôtel-Dieu de Québec (CHUQ); Quebec City, QC Canada
| | - Jacques Côté
- Laval University Cancer Research Center; Hôtel-Dieu de Québec (CHUQ); Quebec City, QC Canada
| | - Xiang-Jiao Yang
- Rosalind and Morris Goodman Cancer Research Center; Departments of Medicine, Biochemistry, and Anatomy & Cell Biology; McGill University; Montréal, QC Canada
| | - Tatiana G Kutateladze
- Department of Pharmacology; University of Colorado School of Medicine; Aurora, CO USA
| |
Collapse
|
5
|
Jafarnejad SM, Li G. Regulation of p53 by ING family members in suppression of tumor initiation and progression. Cancer Metastasis Rev 2012; 31:55-73. [PMID: 22095030 DOI: 10.1007/s10555-011-9329-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The INhibitor of Growth (ING) family is an evolutionarily conserved set of proteins, implicated in suppression of initiation and progression of cancers in various tissues. They promote cell cycle arrest, cellular senescence and apoptosis, participate in stress responses, regulate DNA replication and DNA damage responses, and inhibit cancer cell migration, invasion, and angiogenesis of the tumors. At the molecular level, ING proteins are believed to participate in chromatin remodeling and transcriptional regulation of their target genes. However, the best known function of ING proteins is their cooperation with p53 tumor suppressor protein in tumor suppression. All major isoforms of ING family members can promote the transactivition of p53 and the majority of them are shown to directly interact with p53. In addition, ING proteins are thought to interact with and modulate the function of auxiliary members of p53 pathway, such as MDM2, ARF , p300, and p21, indicating their widespread involvement in the regulation and function of this prominent tumor suppressor pathway. It seems that p53 pathway is the main mechanism by which ING proteins exert their functions. Nevertheless, regulation of other pathways which are not relevant to p53, yet important for tumorigenesis such as TGF-β and NF-κB, by ING proteins is also observed. This review summarizes the current understanding of the mutual interactions and cooperation between different members of ING family with p53 pathway and implications of this cooperation in the suppression of cancer initiation and progression.
Collapse
Affiliation(s)
- Seyed Mehdi Jafarnejad
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
6
|
Conserved molecular interactions within the HBO1 acetyltransferase complexes regulate cell proliferation. Mol Cell Biol 2011; 32:689-703. [PMID: 22144582 DOI: 10.1128/mcb.06455-11] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Acetyltransferase complexes of the MYST family with distinct substrate specificities and functions maintain a conserved association with different ING tumor suppressor proteins. ING complexes containing the HBO1 acetylase are a major source of histone H3 and H4 acetylation in vivo and play critical roles in gene regulation and DNA replication. Here, our molecular dissection of HBO1/ING complexes unravels the protein domains required for their assembly and function. Multiple PHD finger domains present in different subunits bind the histone H3 N-terminal tail with a distinct specificity toward lysine 4 methylation status. We show that natively regulated association of the ING4/5 PHD domain with HBO1-JADE determines the growth inhibitory function of the complex, linked to its tumor suppressor activity. Functional genomic analyses indicate that the p53 pathway is a main target of the complex, at least in part through direct transcription regulation at the initiation site of p21/CDKN1A. These results demonstrate the importance of ING association with MYST acetyltransferases in controlling cell proliferation, a regulated link that accounts for the reported tumor suppressor activities of these complexes.
Collapse
|
7
|
Kiok K, Sun H, Clancy H, Bose S, Kluz T, Wu F, Costa M. Liprin-α4 is required for nickel induced receptor protein tyrosine phosphatase-leukocyte antigen related receptor F (RPTP-LAR) activity. PLoS One 2011; 6:e22764. [PMID: 21829649 PMCID: PMC3150438 DOI: 10.1371/journal.pone.0022764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 06/29/2011] [Indexed: 01/04/2023] Open
Abstract
Liprin-α4 was strongly induced following nickel (II) chloride exposure in a variety of cell types including BEAS-2B, A549, BEP2D and BL41 cells. Liprin-α4, a member of the Liprin alpha family, has seven isoforms but only three of these variants were detected in BEAS-2B cells (004, 201 and 202). The level of Liprin-α4 variants 201 and 004 were highly increased in BEAS-2B cells in response to nickel. We showed that Liprin-α4 bound directly to the cytoplasmic region of RPTP-LAR (receptor protein tyrosine phosphatase-leukocyte antigen-related receptor F). The cytoplasmic region of RPTP-LAR contains two phosphatase domains but only the first domain shows activity. The second domain interacts with other proteins. The phosphatase activity was increased both following nickel treatment and also in the presence of nickel ions in cell extracts. Liprin-α4 knock-down lines with decreased expression of Liprin-α4 variants 004 and 201 exhibited greater nickel toxicity compared to controls. The RPTP-LAR phosphatase activity was only slightly increased in a Liprin-α4 knock-down line. Liprin-α4 appeared necessary for the nickel induced tyrosine phosphatase activity. The presence of Liprin-α4 and nickel increased tyrosine phosphatase activity that reduced the global levels of tyrosine phosphorylation in the cell.
Collapse
Affiliation(s)
- Kathrin Kiok
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Hong Sun
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Hailey Clancy
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Sutapa Bose
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Thomas Kluz
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Fen Wu
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
- * E-mail:
| |
Collapse
|
8
|
Abstract
The lack of effective conventional therapies for the treatment of advanced stage melanoma has stimulated interest in the development of novel strategies for the management of patients with malignant melanoma. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma. For many years, T cell-based immunotherapy has been emphasized in part because of the disappointing results of the monoclonal antibody (mAb)-based clinical trials conducted in the early 1980s and in part because of the postulated major role played by T cells in tumor growth control. More recently, mAb-based therapies have gained in popularity given their clinical and commercial success for a variety of malignant diseases. As a result, there has been increased interest in identifying and characterizing antibody-defined melanoma antigens. Among them, the chondroitin sulfate proteoglycan 4 (CSPG4), also known as high molecular weight-melanoma associated antigen (HMW-MAA) or melanoma chondroitin sulfate proteoglycan (MCSP), has attracted much attention in recent years because of the growing experimental evidence that it fulfills two requirements for immunotherapy to be therapeutically effective: (1) targeting of cancer stem cells (CSC) and (2) development of combinatorial therapies to counteract the escape mechanisms driven by the genetic instability of tumor cells. With this in mind, in this chapter, we have reviewed recent information related to the distribution of CSPG4 on various types of tumors, including CSC, its expression on pericytes in the tumor microenvironment, its recognition by T cells, its role in cell biology as well as the potential mechanisms underlying the ability of CSPG4-specific immunity to control malignant cell growth.
Collapse
|
9
|
Aguissa-Touré AH, Wong RPC, Li G. The ING family tumor suppressors: from structure to function. Cell Mol Life Sci 2011; 68:45-54. [PMID: 20803232 PMCID: PMC11114739 DOI: 10.1007/s00018-010-0509-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 07/31/2010] [Accepted: 08/10/2010] [Indexed: 12/24/2022]
Abstract
The Inhibitor of Growth (ING) proteins belong to a well-conserved family which presents in diverse organisms with several structural and functional domains for each protein. The ING family members are found in association with many cellular processes. Thus, the ING family proteins are involved in regulation of gene transcription, DNA repair, tumorigenesis, apoptosis, cellular senescence and cell cycle arrest. The ING proteins have multiple domains that are potentially capable of binding to many partners. It is conceivable, therefore, that such proteins could function similarly within protein complexes. In this case, within this family, each function could be attributed to a specific domain. However, the role of ING domains is not definitively clear. In this review, we summarize recent advances in structure-function relationships in ING proteins. For each domain, we describe the known biological functions and the approaches utilized to identify the functions associated with ING proteins.
Collapse
Affiliation(s)
- Almass-Houd Aguissa-Touré
- Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Ronald P. C. Wong
- Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Gang Li
- Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| |
Collapse
|