1
|
Herrmann D, Meng S, Yang H, Mansky LM, Saad JS. The Assembly of HTLV-1-How Does It Differ from HIV-1? Viruses 2024; 16:1528. [PMID: 39459862 PMCID: PMC11512237 DOI: 10.3390/v16101528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Retroviral assembly is a highly coordinated step in the replication cycle. The process is initiated when the newly synthesized Gag and Gag-Pol polyproteins are directed to the inner leaflet of the plasma membrane (PM), where they facilitate the budding and release of immature viral particles. Extensive research over the years has provided crucial insights into the molecular determinants of this assembly step. It is established that Gag targeting and binding to the PM is mediated by interactions of the matrix (MA) domain and acidic phospholipids such as phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). This binding event, along with binding to viral RNA, initiates oligomerization of Gag on the PM, a process mediated by the capsid (CA) domain. Much of the previous studies have focused on human immunodeficiency virus type 1 (HIV-1). Although the general steps of retroviral replication are consistent across different retroviruses, comparative studies revealed notable differences in the structure and function of viral components. In this review, we present recent findings on the assembly mechanisms of Human T-cell leukemia virus type 1 and highlight key differences from HIV-1, focusing particularly on the molecular determinants of Gag-PM interactions and CA assembly.
Collapse
Affiliation(s)
- Dominik Herrmann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Shuyu Meng
- Institute for Molecular Virology, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA; (S.M.); (H.Y.); (L.M.M.)
- Molecular Pharmacology and Therapeutics Graduate Program, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA
| | - Huixin Yang
- Institute for Molecular Virology, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA; (S.M.); (H.Y.); (L.M.M.)
| | - Louis M. Mansky
- Institute for Molecular Virology, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA; (S.M.); (H.Y.); (L.M.M.)
- Molecular Pharmacology and Therapeutics Graduate Program, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA
- Department of Diagnostic and Biological Sciences, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota–Twin Cities, Minneapolis, MN 55455, USA
| | - Jamil S. Saad
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
2
|
Lopez A, Nichols Doyle R, Sandoval C, Nisson K, Yang V, Fregoso OI. Viral Modulation of the DNA Damage Response and Innate Immunity: Two Sides of the Same Coin. J Mol Biol 2022; 434:167327. [PMID: 34695379 PMCID: PMC9119581 DOI: 10.1016/j.jmb.2021.167327] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022]
Abstract
The DDR consists of multiple pathways that sense, signal, and respond to anomalous DNA. To promote efficient replication, viruses have evolved to engage and even modulate the DDR. In this review, we will discuss a select set of diverse viruses and the range of mechanisms they evolved to interact with the DDR and some of the subsequent cellular consequences. There is a dichotomy in that the DDR can be both beneficial for viruses yet antiviral. We will also review the connection between the DDR and innate immunity. Previously believed to be disparate cellular functions, more recent research is emerging that links these processes. Furthermore, we will discuss some discrepancies in the literature that we propose can be remedied by utilizing more consistent DDR-focused assays. By doing so, we hope to obtain a much clearer understanding of how broadly these mechanisms and phenotypes are conserved among all viruses. This is crucial for human health since understanding how viruses manipulate the DDR presents an important and tractable target for antiviral therapies.
Collapse
Affiliation(s)
- Andrew Lopez
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Randilea Nichols Doyle
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Carina Sandoval
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Karly Nisson
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Vivian Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Oliver I Fregoso
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
3
|
He Y, Pasupala N, Zhi H, Dorjbal B, Hussain I, Shih HM, Bhattacharyya S, Biswas R, Miljkovic M, Semmes OJ, Waldmann TA, Snow AL, Giam CZ. NF-κB-induced R-loop accumulation and DNA damage select for nucleotide excision repair deficiencies in adult T cell leukemia. Proc Natl Acad Sci U S A 2021; 118:e2005568118. [PMID: 33649200 PMCID: PMC7958262 DOI: 10.1073/pnas.2005568118] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Constitutive NF-κB activation (NF-κBCA) confers survival and proliferation advantages to cancer cells and frequently occurs in T/B cell malignancies including adult T cell leukemia (ATL) caused by human T-cell leukemia virus type 1 (HTLV-1). Counterintuitively, NF-κBCA by the HTLV-1 transactivator/oncoprotein Tax induces a senescence response, and HTLV-1 infections in culture mostly result in senescence or cell-cycle arrest due to NF-κBCA How NF-κBCA induces senescence, and how ATL cells maintain NF-κBCA and avert senescence, remain unclear. Here we report that NF-κBCA by Tax increases R-loop accumulation and DNA double-strand breaks, leading to senescence. R-loop reduction via RNase H1 overexpression, and short hairpin RNA silencing of two transcription-coupled nucleotide excision repair (TC-NER) endonucleases that are critical for R-loop excision-Xeroderma pigmentosum F (XPF) and XPG-attenuate Tax senescence, enabling HTLV-1-infected cells to proliferate. Our data indicate that ATL cells are often deficient in XPF, XPG, or both and are hypersensitive to ultraviolet irradiation. This TC-NER deficiency is found in all ATL types. Finally, ATL cells accumulate R-loops in abundance. Thus, TC-NER deficits are positively selected during HTLV-1 infection because they facilitate the outgrowth of infected cells initially and aid the proliferation of ATL cells with NF-κBCA later. We suggest that TC-NER deficits and excess R-loop accumulation represent specific vulnerabilities that may be targeted for ATL treatment.
Collapse
Affiliation(s)
- Yunlong He
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Nagesh Pasupala
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Huijun Zhi
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Batsuhk Dorjbal
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Imran Hussain
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Hsiu-Ming Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County 350, Taiwan
| | - Sharmistha Bhattacharyya
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Roopa Biswas
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Milos Miljkovic
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Oliver John Semmes
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501
- The Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23501
| | - Thomas A Waldmann
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Chou-Zen Giam
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814;
| |
Collapse
|
4
|
Salamango DJ, Harris RS. Dual Functionality of HIV-1 Vif in APOBEC3 Counteraction and Cell Cycle Arrest. Front Microbiol 2021; 11:622012. [PMID: 33510734 PMCID: PMC7835321 DOI: 10.3389/fmicb.2020.622012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/11/2020] [Indexed: 01/02/2023] Open
Abstract
Accessory proteins are a key feature that distinguishes primate immunodeficiency viruses such as human immunodeficiency virus type I (HIV-1) from other retroviruses. A prime example is the virion infectivity factor, Vif, which hijacks a cellular co-transcription factor (CBF-β) to recruit a ubiquitin ligase complex (CRL5) to bind and degrade antiviral APOBEC3 enzymes including APOBEC3D (A3D), APOBEC3F (A3F), APOBEC3G (A3G), and APOBEC3H (A3H). Although APOBEC3 antagonism is essential for viral pathogenesis, and a more than sufficient functional justification for Vif’s evolution, most viral proteins have evolved multiple functions. Indeed, Vif has long been known to trigger cell cycle arrest and recent studies have shed light on the underlying molecular mechanism. Vif accomplishes this function using the same CBF-β/CRL5 ubiquitin ligase complex to degrade a family of PPP2R5 phospho-regulatory proteins. These advances have helped usher in a new era of accessory protein research and fresh opportunities for drug development.
Collapse
Affiliation(s)
- Daniel J Salamango
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,Institute for Molecular Virology, University of Minnesota, Minneapolis, MN, United States
| | - Reuben S Harris
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,Institute for Molecular Virology, University of Minnesota, Minneapolis, MN, United States.,Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
5
|
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) was discovered in 1980 as the first, and to date, the only retrovirus that causes human cancer. While HTLV-1 infection is generally asymptomatic, 3-5% of infected individuals develop a T cell neoplasm known as adult T cell leukemia/lymphoma (ATL) decades after infection. Since its discovery, HTLV-1 has served as a model for understanding retroviral oncogenesis, transcriptional regulation, cellular signal transduction, and cell-associated viral infection and spread. Much of the initial research was focused on the viral trans-activator/oncoprotein, Tax. Over the past decade, the study of HTLV-1 has entered the genomic era. With the development of new systems for studying HTLV-1 infection and pathogenesis, the completion of the whole genome, exome and transcriptome sequencing analyses of ATL, and the discovery of HBZ as another HTLV-1 oncogene, many established concepts about how HTLV-1 infects, persists and causes disease have undergone substantial revision. This chapter seeks to integrate our current understanding of the mechanisms of action of Tax and HBZ with the comprehensive genomic information of ATL to provide an overview of how HTLV-1 infects, replicates and causes leukemia.
Collapse
|
6
|
Zhi H, Guo X, Ho YK, Pasupala N, Engstrom HAA, Semmes OJ, Giam CZ. RNF8 Dysregulation and Down-regulation During HTLV-1 Infection Promote Genomic Instability in Adult T-Cell Leukemia. PLoS Pathog 2020; 16:e1008618. [PMID: 32453758 PMCID: PMC7274470 DOI: 10.1371/journal.ppat.1008618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/05/2020] [Accepted: 05/11/2020] [Indexed: 12/22/2022] Open
Abstract
The genomic instability associated with adult T cell leukemia/lymphoma (ATL) is causally linked to Tax, the HTLV-1 viral oncoprotein, but the underlying mechanism is not fully understood. We have previously shown that Tax hijacks and aberrantly activates ring finger protein 8 (RNF8) — a lysine 63 (K63)-specific ubiquitin E3 ligase critical for DNA double-strand break (DSB) repair signaling — to assemble K63-linked polyubiquitin chains (K63-pUbs) in the cytosol. Tax and the cytosolic K63-pUbs, in turn, initiate additional recruitment of linear ubiquitin assembly complex (LUBAC) to produce hybrid K63-M1 pUbs, which trigger a kinase cascade that leads to canonical IKK:NF-κB activation. Here we demonstrate that HTLV-1-infected cells are impaired in DNA damage response (DDR). This impairment correlates with the induction of microscopically visible nuclear speckles by Tax known as the Tax-speckle structures (TSS), which act as pseudo DNA damage signaling scaffolds that sequester DDR factors such as BRCA1, DNA-PK, and MDC1. We show that TSS co-localize with Tax, RNF8 and K63-pUbs, and their formation depends on RNF8. Tax mutants defective or attenuated in inducing K63-pUb assembly are deficient or tempered in TSS induction and DDR impairment. Finally, our results indicate that loss of RNF8 expression reduces HTLV-1 viral gene expression and frequently occurs in ATL cells. Thus, during HTLV-1 infection, Tax activates RNF8 to assemble nuclear K63-pUbs that sequester DDR factors in Tax speckles, disrupting DDR signaling and DSB repair. Down-regulation of RNF8 expression is positively selected during infection and progression to disease, and further exacerbates the genomic instability of ATL. Approximately 3–5% of HTLV-1-infected individuals develop an intractable malignancy called adult T cell leukemia/lymphoma (ATL) decades after infection. Unlike other leukemia, ATL is characterized by extensive genomic instability. Here we show that the genomic instability of ATL is associated with the hijacking and aberrant activation of a molecule known as ring finger protein 8 (RNF8) by HTLV-1 for viral replication. RNF8 is crucial for initiating the cellular DNA damage response (DDR) required for the repair of DNA double-strand breaks (DSBs), the most deleterious DNA damage. Its dysregulation in HTLV-1-infected cells results in the formation of pseudo DNA damage signaling scaffolds known as Tax speckle structures that sequester critical repair factors, causing an inability to repair DSBs efficiently. We have further found that loss of RNF8 expression reduces HTLV-1 viral replication and frequently occurs in ATL of all types. This likely facilitates the immune evasion of virus-infected cells, but degrades their ability to repair DSBs and exacerbates the genomic instability of ATL cells. Since DDR defects impact cancer response to DNA-damaging radiation and chemotherapies, RNF8 deficiency in ATL may be exploited for disease treatment.
Collapse
Affiliation(s)
- Huijun Zhi
- Department of Microbiology and Immunology Uniformed Services University of the Health Sciences Bethesda, MD, United States of America
| | - Xin Guo
- Department of Microbiology and Molecular Cell Biology The Leroy T. Canoles Jr Cancer Research Center Eastern Virginia Medical School Norfolk, VA, United States of America
| | - Yik-Khuan Ho
- Department of Microbiology and Immunology Uniformed Services University of the Health Sciences Bethesda, MD, United States of America
| | - Nagesh Pasupala
- Department of Microbiology and Immunology Uniformed Services University of the Health Sciences Bethesda, MD, United States of America
| | - Hampus Alexander Anders Engstrom
- Department of Microbiology and Molecular Cell Biology The Leroy T. Canoles Jr Cancer Research Center Eastern Virginia Medical School Norfolk, VA, United States of America
| | - Oliver John Semmes
- Department of Microbiology and Molecular Cell Biology The Leroy T. Canoles Jr Cancer Research Center Eastern Virginia Medical School Norfolk, VA, United States of America
- * E-mail: (OJS); (C-ZG)
| | - Chou-Zen Giam
- Department of Microbiology and Immunology Uniformed Services University of the Health Sciences Bethesda, MD, United States of America
- * E-mail: (OJS); (C-ZG)
| |
Collapse
|
7
|
Nakamura-Hoshi M, Suzuki T, Ainai A, Hasegawa H, Ishii H, Matano T. Inefficient Tax-Specific T-Cell Responses in Mice after Syngeneic Transplantation with tax-Transgenic Mouse-Derived Adult T-Cell Leukemia Cells. Jpn J Infect Dis 2020; 73:221-225. [PMID: 32009059 DOI: 10.7883/yoken.jjid.2019.494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Adult T-cell leukemia (ATL) is induced by chronic latent infection with human T-cell leukemia virus type 1 (HTLV-1). HTLV-1 Tax is an oncogenic factor that can be targeted by host T-cell responses. However, the expression of Tax in vivo is little in ATL cells and the impact of Tax-specific T-cell responses on ATL progression remains unclear. In the present study, we examined Tax-specific T-cell responses in C57BL/6 mice after syngeneic transplantation with tax-transgenic mouse-derived ATL (mATL) cells. We first confirmed that cellular tax cDNAs are mostly maintained and detectable in the spleen three weeks after mATL cell transplantation. However, mATL cell transplantation did not induce significant Tax-specific T-cell responses. Mice immunized with DNA and adenovirus vectors expressing Tax exhibited Tax-specific CD4+ T-cell responses but showed no enhancement of the responses or reduction in cellular tax cDNA levels after mATL cell transplantation. This study provides an animal model for analyzing the interaction between ATL cells and host immune responses as well as indicates the limited impact of Tax-specific T-cell responses on the proliferation of ATL cells.
Collapse
Affiliation(s)
| | | | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases.,Global Virus Network
| | - Hiroshi Ishii
- AIDS Research Center, National Institute of Infectious Diseases
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases.,The Institute of Medical Science, The University of Tokyo.,Joint Research Center for Human Retrovirus Infection, Kumamoto University
| |
Collapse
|
8
|
The human T-cell leukemia virus type-1 tax oncoprotein dissociates NF-κB p65 RelA-Stathmin complexes and causes catastrophic mitotic spindle damage and genomic instability. Virology 2019; 535:83-101. [PMID: 31299491 DOI: 10.1016/j.virol.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 12/23/2022]
Abstract
Genomic instability is a hallmark of many cancers; however, the molecular etiology of chromosomal dysregulation is not well understood. The human T-cell leukemia virus type-1 (HTLV-1) oncoprotein Tax activates NF-κB-signaling and induces DNA-damage and aberrant chromosomal segregation through diverse mechanisms which contribute to viral carcinogenesis. Intriguingly, Stathmin/oncoprotein-18 (Op-18) depolymerizes tubulin and interacts with the p65RelA subunit and functions as a cofactor for NF-κB-dependent transactivation. We thus hypothesized that the dissociation of p65RelA-Stathmin/Op-18 complexes by Tax could lead to the catastrophic destabilization of microtubule (MT) spindle fibers during mitosis and provide a novel mechanistic link between NF-κB-signaling and genomic instability. Here we report that the inhibition of Stathmin expression by the retroviral latency protein, p30II, or knockdown with siRNA-stathmin, dampens Tax-mediated NF-κB transactivation and counters Tax-induced genomic instability and cytotoxicity. The Tax-G148V mutant, defective for NF-κB activation, exhibited reduced p65RelA-Stathmin binding and diminished genomic instability and cytotoxicity. Dominant-negative inhibitors of NF-κB also prevented Tax-induced multinucleation and apoptosis. Moreover, cell clones containing the infectious HTLV-1 ACH. p30II mutant provirus, impaired for p30II production, exhibited increased multinucleation and the accumulation of cytoplasmic tubulin aggregates following nocodozole-treatment. These findings allude to a mechanism whereby NF-κB-signaling regulates tubulin dynamics and mitotic instability through the modulation of p65RelA-Stathmin/Op-18 interactions, and support the notion that p30II enhances the survival of Tax-expressing HTLV-1-transformed cells.
Collapse
|
9
|
Fan Y, Sanyal S, Bruzzone R. Breaking Bad: How Viruses Subvert the Cell Cycle. Front Cell Infect Microbiol 2018; 8:396. [PMID: 30510918 PMCID: PMC6252338 DOI: 10.3389/fcimb.2018.00396] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/22/2018] [Indexed: 01/10/2023] Open
Abstract
Interactions between the host and viruses during the course of their co-evolution have not only shaped cellular function and the immune system, but also the counter measures employed by viruses. Relatively small genomes and high replication rates allow viruses to accumulate mutations and continuously present the host with new challenges. It is therefore, no surprise that they either escape detection or modulate host physiology, often by redirecting normal cellular pathways to their own advantage. Viruses utilize a diverse array of strategies and molecular targets to subvert host cellular processes, while evading detection. These include cell-cycle regulation, major histocompatibility complex-restricted antigen presentation, intracellular protein transport, apoptosis, cytokine-mediated signaling, and humoral immune responses. Moreover, viruses routinely manipulate the host cell cycle to create a favorable environment for replication, largely by deregulating cell cycle checkpoints. This review focuses on our current understanding of the molecular aspects of cell cycle regulation that are often targeted by viruses. Further study of their interactions should provide fundamental insights into cell cycle regulation and improve our ability to exploit these viruses.
Collapse
Affiliation(s)
- Ying Fan
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sumana Sanyal
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,LKS Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong, Hong Kong
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| |
Collapse
|
10
|
Yamagishi M, Fujikawa D, Watanabe T, Uchimaru K. HTLV-1-Mediated Epigenetic Pathway to Adult T-Cell Leukemia-Lymphoma. Front Microbiol 2018; 9:1686. [PMID: 30087673 PMCID: PMC6066519 DOI: 10.3389/fmicb.2018.01686] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/06/2018] [Indexed: 11/13/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1), the first reported human oncogenic retrovirus, is the etiologic agent of highly aggressive, currently incurable diseases such as adult T-cell leukemia-lymphoma (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). HTLV-1 proteins, including Tax and HBZ, have been shown to have critical roles in HTLV-1 pathogenicity, yet the underlying mechanisms of HTLV-1-driven leukemogenesis are unclear. The frequent disruption of genetic and epigenetic gene regulation in various types of malignancy, including ATL, is evident. In this review, we illustrate a focused range of topics about the establishment of HTLV-1 memory: (1) genetic lesion in the Tax interactome pathway, (2) gene regulatory loop/switch, (3) disordered chromatin regulation, (4) epigenetic lock by the modulation of epigenetic factors, (5) the loss of gene fine-tuner microRNA, and (6) the alteration of chromatin regulation by HTLV-1 integration. We discuss the persistent influence of Tax-dependent epigenetic changes even after the disappearance of HTLV-1 gene expression due to the viral escape from the immune system, which is a remaining challenge in HTLV-1 research. The summarized evidence and conceptualized description may provide a better understanding of HTLV-1-mediated cellular transformation and the potential therapeutic strategies to combat HTLV-1-associated diseases.
Collapse
Affiliation(s)
- Makoto Yamagishi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Dai Fujikawa
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiki Watanabe
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kaoru Uchimaru
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Giam CZ, Semmes OJ. HTLV-1 Infection and Adult T-Cell Leukemia/Lymphoma-A Tale of Two Proteins: Tax and HBZ. Viruses 2016; 8:v8060161. [PMID: 27322308 PMCID: PMC4926181 DOI: 10.3390/v8060161] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/19/2022] Open
Abstract
HTLV-1 (Human T-cell lymphotropic virus type 1) is a complex human delta retrovirus that currently infects 10–20 million people worldwide. While HTLV-1 infection is generally asymptomatic, 3%–5% of infected individuals develop a highly malignant and intractable T-cell neoplasm known as adult T-cell leukemia/lymphoma (ATL) decades after infection. How HTLV-1 infection progresses to ATL is not well understood. Two viral regulatory proteins, Tax and HTLV-1 basic zipper protein (HBZ), encoded by the sense and antisense viral transcripts, respectively, are thought to play indispensable roles in the oncogenic process of ATL. This review focuses on the roles of Tax and HBZ in viral replication, persistence, and oncogenesis. Special emphasis is directed towards recent literature on the mechanisms of action of these two proteins and the roles of Tax and HBZ in influencing the outcomes of HTLV-1 infection including senescence induction, viral latency and persistence, genome instability, cell proliferation, and ATL development. Attempts are made to integrate results from cell-based studies of HTLV-1 infection and studies of HTLV-1 proviral integration site preference, clonality, and clonal expansion based on high throughput DNA sequencing. Recent data showing that Tax hijacks key mediators of DNA double-strand break repair signaling—the ubiquitin E3 ligase, ring finger protein 8 (RNF8) and the ubiquitin E2 conjugating enzyme (UBC13)—to activate the canonical nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) and other signaling pathways will be discussed. A perspective on how the Tax-RNF8 signaling axis might impact genomic instability and how Tax may collaborate with HBZ to drive oncogenesis is provided.
Collapse
Affiliation(s)
- Chou-Zen Giam
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA.
| | - Oliver John Semmes
- Department of Microbiology and Molecular Cell Biology, The Leroy T. Canoles Jr Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23501, USA.
| |
Collapse
|
12
|
Nakano K, Watanabe T. HTLV-1 Rex Tunes the Cellular Environment Favorable for Viral Replication. Viruses 2016; 8:58. [PMID: 26927155 PMCID: PMC4810248 DOI: 10.3390/v8030058] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/09/2016] [Accepted: 02/09/2016] [Indexed: 12/18/2022] Open
Abstract
Human T-cell leukemia virus type-1 (HTLV-1) Rex is a viral RNA binding protein. The most important and well-known function of Rex is stabilizing and exporting viral mRNAs from the nucleus, particularly for unspliced/partially-spliced mRNAs encoding the structural proteins essential for viral replication. Without Rex, these unspliced viral mRNAs would otherwise be completely spliced. Therefore, Rex is vital for the translation of structural proteins and the stabilization of viral genomic RNA and, thus, for viral replication. Rex schedules the period of extensive viral replication and suppression to enter latency. Although the importance of Rex in the viral life-cycle is well understood, the underlying molecular mechanism of how Rex achieves its function has not been clarified. For example, how does Rex protect unspliced/partially-spliced viral mRNAs from the host cellular splicing machinery? How does Rex protect viral mRNAs, antigenic to eukaryotic cells, from cellular mRNA surveillance mechanisms? Here we will discuss these mechanisms, which explain the function of Rex as an organizer of HTLV-1 expression based on previously and recently discovered aspects of Rex. We also focus on the potential influence of Rex on the homeostasis of the infected cell and how it can exert its function.
Collapse
Affiliation(s)
- Kazumi Nakano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 4-6-1, Shirokanedai, Minatoku, Tokyo 108-8639, Japan.
| | - Toshiki Watanabe
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 4-6-1, Shirokanedai, Minatoku, Tokyo 108-8639, Japan.
| |
Collapse
|
13
|
Chen L, Liu D, Zhang Y, Zhang H, Cheng H. Foxp3-dependent transformation of human primary CD4+ T lymphocytes by the retroviral protein tax. Biochem Biophys Res Commun 2015; 466:523-9. [PMID: 26381169 DOI: 10.1016/j.bbrc.2015.09.063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/11/2015] [Indexed: 12/31/2022]
Abstract
The retroviral Tax proteins of human T cell leukemia virus type 1 and 2 (HTLV-1 and -2) are highly homologous viral transactivators. Both viral proteins can immortalize human primary CD4+ memory T cells, but when expressed alone they rarely transform T cells. In the present study, we found that the Tax proteins displayed a differential ability to immortalize human CD4+Foxp3+ T cells with characteristic expression of CTLA-4 and GITR. Because epidermal growth factor receptor (EGFR) was reportedly expressed and activated in a subset of CD4+Foxp3+ T cells, we introduced an activated EGFR into Tax-immortalized CD4+Foxp3+ T cells. We observed that these modified cells were grown independently of exogenous IL-2, correlating with a T cell transformation phenotype. In Tax-immortalized CD4+Foxp3- T cells, ectopic expression of Foxp3 was a prerequisite for Tax transformation of T cells. Accordingly, treatment of the transformed T cells with erlotinib, a selective inhibitor of EGFR, induced degradation of EGFR in lysosome, consequently causing T cell growth inhibition. Further, we identified autophagy as a crucial cellular survival pathway for the transformed T cells. Silencing key autophagy molecules including Beclin1, Atg5 and PI3 kinase class III (PI3KC3) resulted in drastic impairment of T cell growth. Our data, therefore, unveiled a previously unidentified role of Foxp3 in T cell transformation, providing a molecular basis for HTLV-1 transformation of CD4+Foxp3+ T cells.
Collapse
Affiliation(s)
- Li Chen
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dan Liu
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yang Zhang
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Huan Zhang
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hua Cheng
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Departments of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Departments of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
14
|
Buehring GC, Shen HM, Jensen HM, Jin DL, Hudes M, Block G. Exposure to Bovine Leukemia Virus Is Associated with Breast Cancer: A Case-Control Study. PLoS One 2015; 10:e0134304. [PMID: 26332838 PMCID: PMC4557937 DOI: 10.1371/journal.pone.0134304] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/07/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Age, reproductive history, hormones, genetics, and lifestyle are known risk factors for breast cancer, but the agents that initiate cellular changes from normal to malignant are not understood. We previously detected bovine leukemia virus (BLV), a common oncogenic virus of cattle, in the breast epithelium of humans. The objective of this study was to determine whether the presence of BLV DNA in human mammary epithelium is associated with breast cancer. METHODS This was a case-control study of archival formalin fixed paraffin embedded breast tissues from 239 donors, received 2002-2008 from the Cooperative Human Tissue Network. Case definition as breast cancer versus normal (women with no history of breast cancer) was established through medical records and examination of tissues by an anatomical pathologist. Breast exposure to BLV was determined by in situ-PCR detection of a biomarker, BLV DNA, localized within mammary epithelium. RESULTS The frequency of BLV DNA in mammary epithelium from women with breast cancer (59%) was significantly higher than in normal controls (29%) (multiply- adjusted odds ratio = 3.07, confidence interval = 1.66-5.69, p = .0004, attributable risk = 37%). In women with premalignant breast changes the frequency of BLV DNA was intermediate (38%) between that of women with breast cancer and normal controls (p for trend < .001). CONCLUSIONS Among the specimens in this study, the presence of amplified BLV DNA was significantly associated with breast cancer. The odds ratio magnitude was comparable to those of well-established breast cancer risk factors related to reproductive history, hormones, and lifestyle and was exceeded only by risk factors related to genetics (familial breast cancer), high dose ionizing radiation, and age. These findings have the potential for primary and secondary prevention of breast cancer.
Collapse
Affiliation(s)
- Gertrude Case Buehring
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United States of America
- * E-mail:
| | - Hua Min Shen
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United States of America
| | - Hanne M. Jensen
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, California, United States of America
| | - Diana L. Jin
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United States of America
| | - Mark Hudes
- Atkins Center for Weight and Health, University of California, Berkeley, California, United States of America
| | - Gladys Block
- Division of Community Health and Human Development, School of Public Health, University of California, Berkeley, California, United States of America
| |
Collapse
|
15
|
Modulation of DNA damage and repair pathways by human tumour viruses. Viruses 2015; 7:2542-91. [PMID: 26008701 PMCID: PMC4452920 DOI: 10.3390/v7052542] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/12/2015] [Indexed: 02/07/2023] Open
Abstract
With between 10% and 15% of human cancers attributable to viral infection, there is great interest, from both a scientific and clinical viewpoint, as to how these pathogens modulate host cell functions. Seven human tumour viruses have been identified as being involved in the development of specific malignancies. It has long been known that the introduction of chromosomal aberrations is a common feature of viral infections. Intensive research over the past two decades has subsequently revealed that viruses specifically interact with cellular mechanisms responsible for the recognition and repair of DNA lesions, collectively known as the DNA damage response (DDR). These interactions can involve activation and deactivation of individual DDR pathways as well as the recruitment of specific proteins to sites of viral replication. Since the DDR has evolved to protect the genome from the accumulation of deleterious mutations, deregulation is inevitably associated with an increased risk of tumour formation. This review summarises the current literature regarding the complex relationship between known human tumour viruses and the DDR and aims to shed light on how these interactions can contribute to genomic instability and ultimately the development of human cancers.
Collapse
|
16
|
Kim S, Lee HS, Ji JH, Cho MY, Yoo YS, Park YY, Cha HJ, Lee Y, Kim Y, Cho H. Hepatitis B virus X protein activates the ATM-Chk2 pathway and delays cell cycle progression. J Gen Virol 2015; 96:2242-2251. [PMID: 25872745 DOI: 10.1099/vir.0.000150] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Genetic instability is intimately associated with tumour development. In particular, liver cancers associated with hepatitis B virus (HBV) exhibit high genetic instability; however, our understanding of the underlying molecular mechanisms remains limited. In this study, we found that γ-H2AX, a marker of DNA double-strand breaks (DSBs), and the levels of phospho-Chk2 (p-Chk2, the activated form) were significantly elevated in HBV-associated hepatocellular carcinomas and neighbouring regenerating nodules. Likewise, introduction of the pHBV or pMyc-HBx plasmids into cells induced accumulation of γ-H2AX foci and increased the p-Chk2 level. In these cells, inhibitory phosphorylation of Cdc25C phosphatase (Ser(216)) and CDK1 (Tyr(15)) was elevated; consequently, cell-cycle progression was delayed at G2/M phase, suggesting that activation of the ATM-Chk2 pathway by the HBV X protein (HBx) induces cell-cycle delay. Accordingly, inhibition of ataxia telangiectasia mutated (ATM) by caffeine or siRNA abolished the increase in the p-Chk2 level and restored the delayed CDK1 kinase activity in ChangX cells. We also found that cytoplasmic HBx, but not nuclear HBx, induced reactive oxygen species (ROS) production and led to the accumulation of γ-H2AX foci and the increased p-Chk2 level. Together, these data indicate that HBx-induced ROS accumulation induces DNA damage that activates the ATM-Chk2 pathway. Our findings provide insight into the mechanisms of HBV pathogenesis.
Collapse
Affiliation(s)
- Sujeong Kim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Ho-Soo Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea.,Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon, Republic of Korea
| | - Jae-Hoon Ji
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Mi-Young Cho
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea.,Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon, Republic of Korea
| | - Young-Suk Yoo
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea.,Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon, Republic of Korea
| | - Yong-Yea Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyuk-Jin Cha
- Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| | - Youngsoo Lee
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Youngbae Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyeseong Cho
- Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon, Republic of Korea.,Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea.,Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
17
|
Takachi T, Takahashi M, Takahashi-Yoshita M, Higuchi M, Obata M, Mishima Y, Okuda S, Tanaka Y, Matsuoka M, Saitoh A, Green PL, Fujii M. Human T-cell leukemia virus type 1 Tax oncoprotein represses the expression of the BCL11B tumor suppressor in T-cells. Cancer Sci 2015; 106:461-5. [PMID: 25613934 PMCID: PMC4409891 DOI: 10.1111/cas.12618] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/07/2015] [Accepted: 01/17/2015] [Indexed: 12/31/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T cell leukemia (ATL), which is an aggressive form of T-cell malignancy. HTLV-1 oncoproteins, Tax and HBZ, play crucial roles in the immortalization of T-cells and/or leukemogenesis by dysregulating the cellular functions in the host. Recent studies show that HTLV-1-infected T-cells have reduced expression of the BCL11B tumor suppressor protein. In the present study, we explored whether Tax and/or HBZ play a role in downregulating BCL11B in HTLV-1-infected T-cells. Lentiviral transduction of Tax in a human T-cell line repressed the expression of BCL11B at both the protein and mRNA levels, whereas the transduction of HBZ had little effect on the expression. Tax mutants with a decreased activity for the NF-κB, CREB or PDZ protein pathways still showed a reduced expression of the BCL11B protein, thereby implicating a different function of Tax in BCL11B downregulation. In addition, the HTLV-2 Tax2 protein reduced the BCL11B protein expression in T-cells. Seven HTLV-1-infected T-cell lines, including three ATL-derived cell lines, showed reduced BCL11B mRNA and protein expression relative to an uninfected T-cell line, and the greatest reductions were in the cells expressing Tax. Collectively, these results indicate that Tax is responsible for suppressing BCL11B protein expression in HTLV-1-infected T-cells; Tax-mediated repression of BCL11B is another mechanism that Tax uses to promote oncogenesis of HTLV-1-infected T-cells.
Collapse
Affiliation(s)
- Takayuki Takachi
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Division of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
To replicate their genomes in cells and generate new progeny, viruses typically require factors provided by the cells that they have infected. Subversion of the cellular machinery that controls replication of the infected host cell is a common activity of many viruses. Viruses employ different strategies to deregulate cell cycle checkpoint controls and modulate cell proliferation pathways. A number of DNA and RNA viruses encode proteins that target critical cell cycle regulators to achieve cellular conditions that are beneficial for viral replication. Many DNA viruses induce quiescent cells to enter the cell cycle; this is thought to increase pools of deoxynucleotides and thus, facilitate viral replication. In contrast, some viruses can arrest cells in a particular phase of the cell cycle that is favorable for replication of the specific virus. Cell cycle arrest may inhibit early cell death of infected cells, allow the cells to evade immune defenses, or help promote virus assembly. Although beneficial for the viral life cycle, virus-mediated alterations in normal cell cycle control mechanisms could have detrimental effects on cellular physiology and may ultimately contribute to pathologies associated with the viral infection, including cell transformation and cancer progression and maintenance. In this chapter, we summarize various strategies employed by DNA and RNA viruses to modulate the replication cycle of the virus-infected cell. When known, we describe how these virus-associated effects influence replication of the virus and contribute to diseases associated with infection by that specific virus.
Collapse
Affiliation(s)
- Eishi Noguchi
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania USA
| | - Mariana C. Gadaleta
- Dept of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, USA
| |
Collapse
|
19
|
Lodewick J, Sampaio C, Boxus M, Rinaldi AS, Coulonval K, Willems L, Roger PP, Bex F. Acetylation at lysine 346 controls the transforming activity of the HTLV-1 Tax oncoprotein in the Rat-1 fibroblast model. Retrovirology 2013; 10:75. [PMID: 23880157 PMCID: PMC3734113 DOI: 10.1186/1742-4690-10-75] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/18/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Transformation by the Tax oncoprotein of the human T cell leukemia virus type 1 (HTLV-1) is governed by actions on cellular regulatory signals, including modulation of specific cellular gene expression via activation of signaling pathways, acceleration of cell cycle progression via stimulation of cyclin-dependent kinase activity leading to retinoblastoma protein (pRb) hyperphosphorylation and perturbation of survival signals. These actions control early steps in T cell transformation and development of Adult T cell leukemia (ATL), an aggressive malignancy of HTLV-1 infected T lymphocytes. Post-translational modifications of Tax by phosphorylation, ubiquitination, sumoylation and acetylation have been implicated in Tax-mediated activation of the NF-κB pathway, a key function associated with Tax transforming potential. RESULTS In this study, we demonstrate that acetylation at lysine K(346) in the carboxy-terminal domain of Tax is modulated in the Tax nuclear bodies by the acetyltransferase p300 and the deacetylases HDAC5/7 and controls phosphorylation of the tumor suppressor pRb by Tax-cyclin D3-CDK4-p21(CIP) complexes. This property correlates with the inability of the acetylation deficient K(346)R mutant, but not the acetylation mimetic K(346)Q mutant, to promote anchorage-independent growth of Rat-1 fibroblasts. By contrast, acetylation at lysine K(346) had no effects on the ability of Tax carboxy-terminal PDZ-binding domain to interact with the tumor suppressor hDLG. CONCLUSIONS The identification of the acetyltransferase p300 and the deacetylase HDAC7 as enzymes modulating Tax acetylation points to new therapeutic targets for the treatment of HTLV-1 infected patients at risk of developing ATL.
Collapse
Affiliation(s)
- Julie Lodewick
- Institute for Microbiological Research J-M Wiame (IRMW), Laboratory of Microbiology, Université Libre de Bruxelles, 1, Avenue E, Gryson, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Currer R, Van Duyne R, Jaworski E, Guendel I, Sampey G, Das R, Narayanan A, Kashanchi F. HTLV tax: a fascinating multifunctional co-regulator of viral and cellular pathways. Front Microbiol 2012; 3:406. [PMID: 23226145 PMCID: PMC3510432 DOI: 10.3389/fmicb.2012.00406] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 11/12/2012] [Indexed: 12/18/2022] Open
Abstract
Human T-cell lymphotropic virus type 1 (HTLV-1) has been identified as the causative agent of adult T-cell leukemia (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). The virus infects between 15 and 20 million people worldwide of which approximately 2-5% develop ATL. The past 35 years of research have yielded significant insight into the pathogenesis of HTLV-1, including the molecular characterization of Tax, the viral transactivator, and oncoprotein. In spite of these efforts, the mechanisms of oncogenesis of this pleiotropic protein remain to be fully elucidated. In this review, we illustrate the multiple oncogenic roles of Tax by summarizing a recent body of literature that refines our understanding of cellular transformation. A focused range of topics are discussed in this review including Tax-mediated regulation of the viral promoter and other cellular pathways, particularly the connection of the NF-κB pathway to both post-translational modifications (PTMs) of Tax and subcellular localization. Specifically, recent research on polyubiquitination of Tax as it relates to the activation of the IkappaB kinase (IKK) complex is highlighted. Regulation of the cell cycle and DNA damage responses due to Tax are also discussed, including Tax interaction with minichromosome maintenance proteins and the role of Tax in chromatin remodeling. The recent identification of HTLV-3 has amplified the importance of the characterization of emerging viral pathogens. The challenge of the molecular determination of pathogenicity and malignant disease of this virus lies in the comparison of the viral transactivators of HTLV-1, -2, and -3 in terms of transformation and immortalization. Consequently, differences between the three proteins are currently being studied to determine what factors are required for the differences in tumorogenesis.
Collapse
Affiliation(s)
- Robert Currer
- National Center for Biodefense and Infectious Diseases, George Mason University Manassas, VA, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Cheng H, Ren T, Sun SC. New insight into the oncogenic mechanism of the retroviral oncoprotein Tax. Protein Cell 2012; 3:581-9. [PMID: 22865346 DOI: 10.1007/s13238-012-2047-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/07/2012] [Indexed: 12/29/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1), an etiological factor that causes adult T cell leukemia and lymphoma (ATL), infects over 20 million people worldwide. About 1 million of HTLV-1-infected patients develop ATL, a highly aggressive non-Hodgkin's lymphoma without an effective therapy. The pX region of the HTLV-1 viral genome encodes an oncogenic protein, Tax, which plays a central role in transforming CD4+ T lymphocytes by deregulating oncogenic signaling pathways and promoting cell cycle progression. Expression of Tax following viral entry is critical for promoting survival and proliferation of human T cells and is required for initiation of oncogenesis. Tax exhibits diverse functions in host cells, and this oncoprotein primarily targets IκB kinase complex in the cytoplasm, resulting in persistent activation of NF-κB and upregulation of its responsive gene expressions that are crucial for T cell survival and cell cycle progression. We here review recent advances for the pathological roles of Tax in modulating IκB kinase activity. We also discuss our recent observation that Tax connects the IκB kinase complex to autophagy pathways. Understanding Tax-mediated pathogenesis will provide insights into development of new therapeutics in controlling HTLV-1-associated diseases.
Collapse
Affiliation(s)
- Hua Cheng
- Penn State Hershey Cancer Institute, Hershey, PA 17033, USA.
| | | | | |
Collapse
|
22
|
The multifaceted oncoprotein Tax: subcellular localization, posttranslational modifications, and NF-κB activation. Adv Cancer Res 2012; 113:85-120. [PMID: 22429853 DOI: 10.1016/b978-0-12-394280-7.00003-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The human T-cell lymphotropic virus type-I (HTLV-I) is the etiologic agent of adult T-cell leukemia/lymphoma (ATL) and of tropical spastic paraparesis/HTLV-I-associated myelopathy. Constitutive NF-κB activation by the viral oncoprotein Tax plays a crucial role in the induction and maintenance of cellular proliferation, transformation, and inhibition of apoptosis. In an attempt to provide a general view of the molecular mechanisms of constitutive Tax-induced NF-κB activation, we summarize in this review the recent body of literature that supports a major role for Tax posttranslational modifications, chiefly ubiquitination, and SUMOylation, in the NF-κB activity of Tax. These modifications indeed participate in the control of Tax subcellular localization and modulate its protein-protein interaction potential. Tax posttranslational modifications, which highlight the ability of HTLV-I to optimize its limited viral genome size, might represent an attractive target for the design of new therapies for ATL.
Collapse
|
23
|
Boxus M, Willems L. How the DNA damage response determines the fate of HTLV-1 Tax-expressing cells. Retrovirology 2012; 9:2. [PMID: 22221708 PMCID: PMC3283471 DOI: 10.1186/1742-4690-9-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/05/2012] [Indexed: 11/10/2022] Open
Abstract
How the Human T lymphotropic virus type 1 (HTLV-1) Tax protein stimulates proliferation while triggering cell cycle arrest and senescence remains puzzling. There is also a debate about the ability of Tax to activate or inhibit the DNA damage response. Here, we comment on these different activities and propose a conceptual rationale for the apparently conflicting observations.
Collapse
Affiliation(s)
- Mathieu Boxus
- National Fund for Scientific Research, Gembloux Agro-Bio Tech and Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège, Belgium
| | | |
Collapse
|
24
|
Wang YE, Pernet O, Lee B. Regulation of the nucleocytoplasmic trafficking of viral and cellular proteins by ubiquitin and small ubiquitin-related modifiers. Biol Cell 2011; 104:121-38. [PMID: 22188262 PMCID: PMC3625690 DOI: 10.1111/boc.201100105] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/22/2011] [Indexed: 12/29/2022]
Abstract
Nucleocytoplasmic trafficking of many cellular proteins is regulated by nuclear import/export signals as well as post-translational modifications such as covalent conjugation of ubiquitin and small ubiquitin-related modifiers (SUMOs). Ubiquitination and SUMOylation are rapid and reversible ways to modulate the intracellular localisation and function of substrate proteins. These pathways have been co-opted by some viruses, which depend on the host cell machinery to transport their proteins in and out of the nucleus. In this review, we will summarise our current knowledge on the ubiquitin/SUMO-regulated nuclear/subnuclear trafficking of cellular proteins and describe examples of viral exploitation of these pathways.
Collapse
Affiliation(s)
- Yao E Wang
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
25
|
The Sumo-targeted ubiquitin ligase RNF4 regulates the localization and function of the HTLV-1 oncoprotein Tax. Blood 2011; 119:1173-81. [PMID: 22106342 DOI: 10.1182/blood-2011-06-358564] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The Really Interesting New Gene (RING) Finger Protein 4 (RNF4) represents a class of ubiquitin ligases that target Small Ubiquitin-like Modifier (SUMO)-modified proteins for ubiquitin modification. To date, the regulatory function of RNF4 appears to be ubiquitin-mediated degradation of sumoylated cellular proteins. In the present study, we show that the Human T-cell Leukemia Virus Type 1 (HTLV-1) oncoprotein Tax is a substrate for RNF4 both in vivo and in vitro. We mapped the RNF4-binding site to a region adjacent to the Tax ubiquitin/SUMO modification sites K280/K284. Interestingly, RNF4 modification of Tax protein results in relocalization of the oncoprotein from the nucleus to the cytoplasm. Overexpression of RNF4, but not the RNF4 RING mutant, resulted in cytoplasmic enrichment of Tax. The RNF4-induced nucleus-to-cytoplasm relocalization was associated with increased NF-κB-mediated and decreased cAMP Response Element-Binding (CREB)-mediated Tax activity. Finally, depletion of RNF4 by RNAi prevented the DNA damage-induced nuclear/cytoplasmic translocation of Tax. These results provide important new insight into STUbL-mediated pathways that regulate the subcellular localization and functional dynamics of viral oncogenes.
Collapse
|
26
|
Interaction of HTLV-1 Tax with minichromosome maintenance proteins accelerates the replication timing program. Blood 2011; 119:151-60. [PMID: 22058115 DOI: 10.1182/blood-2011-05-356790] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Tax oncoprotein encoded by the human T-cell leukemia virus type 1 plays a pivotal role in viral persistence and pathogenesis. Human T-cell leukemia virus type 1-infected cells proliferate faster than normal lymphocytes, expand through mitotic division, and accumulate genomic lesions. Here, we show that Tax associates with the minichromosome maintenance MCM2-7 helicase complex and localizes to origins of replication. Tax modulates the spatiotemporal program of origin activation and fires supplementary origins at the onset of S phase. Thereby, Tax increases the DNA replication rate, accelerates S phase progression, but also generates a replicative stress characterized by the presence of genomic lesions. Mechanistically, Tax favors p300 recruitment and histone hyperacetylation at late replication domains, advancing their replication timing in early S phase.
Collapse
|
27
|
Saggioro D. Anti-apoptotic effect of Tax: an NF-κB path or a CREB way? Viruses 2011; 3:1001-14. [PMID: 21994767 PMCID: PMC3185786 DOI: 10.3390/v3071001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/09/2011] [Accepted: 06/11/2011] [Indexed: 12/19/2022] Open
Abstract
The NF-κB pathway is intimately linked to the survival of mammalian cells, and its activation by Tax has consequently been considered important for human T-cell leukemia/lymphoma virus type 1 (HTLV-1)-infected cell resistance to death. Very little emphasis has been given to other mechanisms, although Tax regulates the expression and activity of several cellular genes. The finding that CREB protein is activated in HTLV-1 infected cells underlines the possibility that other mechanisms of survival may be implicated in HTLV-1 infection. Indeed, CREB activation or overexpression plays a role in normal hematopoiesis, as well as in leukemia development, and CREB is considered as a survival factor in various cell systems. A better understanding of the different molecular mechanisms used by Tax to counteract cell death will also help in the development of new therapeutic strategies for HTLV-1 associated diseases.
Collapse
Affiliation(s)
- Daniela Saggioro
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, 35128 Padova, Italy.
| |
Collapse
|
28
|
Lodewick J, Lamsoul I, Bex F. Move or die: the fate of the Tax oncoprotein of HTLV-1. Viruses 2011; 3:829-57. [PMID: 21994756 PMCID: PMC3185767 DOI: 10.3390/v3060829] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Revised: 05/31/2011] [Accepted: 06/01/2011] [Indexed: 12/12/2022] Open
Abstract
The HTLV-1 Tax protein both activates viral replication and is involved in HTLV-1-mediated transformation of T lymphocytes. The transforming properties of Tax include altering the expression of select cellular genes via activation of cellular pathways and perturbation of both cell cycle control mechanisms and apoptotic signals. The recent discovery that Tax undergoes a hierarchical sequence of posttranslational modifications that control its intracellular localization provides provocative insights into the mechanisms regulating Tax transcriptional and transforming activities.
Collapse
Affiliation(s)
- Julie Lodewick
- Institut de Recherches Microbiologiques J-M Wiame, Université Libre de Bruxelles, B-1070 Bruxelles, Belgium.
| | | | | |
Collapse
|
29
|
Legros S, Boxus M, Gatot JS, Van Lint C, Kruys V, Kettmann R, Twizere JC, Dequiedt F. The HTLV-1 Tax protein inhibits formation of stress granules by interacting with histone deacetylase 6. Oncogene 2011; 30:4050-62. [PMID: 21532619 DOI: 10.1038/onc.2011.120] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human T cell leukemia virus type-1 (HTLV-1) is the causative agent of a fatal adult T-cell leukemia. Through deregulation of multiple cellular signaling pathways the viral Tax protein has a pivotal role in T-cell transformation. In response to stressful stimuli, cells mount a cellular stress response to limit the damage that environmental forces inflict on DNA or proteins. During stress response, cells postpone the translation of most cellular mRNAs, which are gathered into cytoplasmic mRNA-silencing foci called stress granules (SGs) and allocate their available resources towards the production of dedicated stress-management proteins. Here we demonstrate that Tax controls the formation of SGs and interferes with the cellular stress response pathway. In agreement with previous reports, we observed that Tax relocates from the nucleus to the cytoplasm in response to environmental stress. We found that the presence of Tax in the cytoplasm of stressed cells prevents the formation of SGs and counteracts the shutoff of specific host proteins. Unexpectedly, nuclear localization of Tax promotes spontaneous aggregation of SGs, even in the absence of stress. Mutant analysis revealed that the SG inhibitory capacity of Tax is independent of its transcriptional abilities but relies on its interaction with histone deacetylase 6, a critical component of SGs. Importantly, the stress-protective effect of Tax was also observed in the context of HTLV-1 infected cells, which were shown to be less prone to form SGs and undergo apoptosis under arsenite exposure. These observations identify Tax as the first virally encoded inhibitory component of SGs and unravel a new strategy developed by HTLV-1 to deregulate normal cell processes. We postulate that inhibition of the stress response pathway by Tax would favor cell survival under stressful conditions and may have an important role in HTLV-1-induced cellular transformation.
Collapse
Affiliation(s)
- S Legros
- Center for Molecular and Cellular Biology, Gembloux Agro-Bio Tech, University of Liège (ULg), Gembloux, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia/lymphoma (ATL), a malignancy of CD4(+) T cells whose etiology is thought to be associated with the viral trans-activator Tax. We have shown recently that Tax can drastically upregulate the expression of p27(Kip1) and p21(CIP1/WAF1) through protein stabilization and mRNA trans-activation and stabilization, respectively. The Tax-induced surge in p21(CIP1/WAF1) and p27(Kip1) begins in S phase and results in cellular senescence. Importantly, HeLa and SupT1 T cells infected by HTLV-1 also arrest in senescence, thus challenging the notion that HTLV-1 infection causes cell proliferation. Here we use time-lapse microscopy to investigate the effect of Tax on cell cycle progression in two reporter cell lines, HeLa/18x21-EGFP and HeLa-FUCCI, that express enhanced green fluorescent protein (EGFP) under the control of 18 copies of the Tax-responsive 21-bp repeat element and fluorescent ubiquitin cell cycle indicators, respectively. Tax-expressing HeLa cells exhibit elongated or stalled cell cycle phases. Many of them bypass mitosis and become single senescent cells as evidenced by the expression of senescence-associated β-galactosidase. Such cells have twice the normal equivalent of cellular contents and hence are enlarged, with exaggerated nuclei. Interestingly, nocodazole treatment revealed a small variant population of HeLa/18x21-EGFP cells that could progress into mitosis normally with high levels of Tax expression, suggesting that genetic or epigenetic changes that prevent Tax-induced senescence can occur spontaneously at a detectable frequency.
Collapse
|
31
|
Belgnaoui SM, Fryrear KA, Nyalwidhe JO, Guo X, Semmes OJ. The viral oncoprotein tax sequesters DNA damage response factors by tethering MDC1 to chromatin. J Biol Chem 2010; 285:32897-32905. [PMID: 20729195 PMCID: PMC2963403 DOI: 10.1074/jbc.m110.146373] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Infection with human T-cell leukemia virus induces cellular genomic instability mediated through the viral oncoprotein Tax. Here we present evidence that Tax undermines the cellular DNA damage response by sequestration of damage response factors. We show by confocal microscopy that Tax forms damage-independent nuclear foci that contain DNA-PK, BRCA1, and MDC1. Tax sequesters MDC1 to chromatin sites distinct from classic ionizing radiation-induced foci. The recruitment of MDC1 is competitive between the two foci. The N-terminal region of Tax is sufficient for foci localization, and the C-terminal half is critical for binding to MDC1 and recruitment of additional response factors. Tax expression and DNA damage response factor recruitment repressed the formation of ionizing radiation-induced Nbs1-containing foci. The Tax-induced “pseudo” DNA damage response results in phosphorylation and monoubiquitylation of H2AX, which is ablated by siRNA suppression of MDC1. These data support a model for virus-induced genomic instability in which viral oncogene-induced damage-independent foci compete with normal cellular DNA damage response.
Collapse
Affiliation(s)
- S Mehdi Belgnaoui
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virgina 23508
| | - Kimberly A Fryrear
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virgina 23508
| | - Julius O Nyalwidhe
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virgina 23508
| | - Xin Guo
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virgina 23508
| | - O John Semmes
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virgina 23508.
| |
Collapse
|
32
|
Guo X, Ward MD, Tiedebohl JB, Oden YM, Nyalwidhe JO, Semmes OJ. Interdependent phosphorylation within the kinase domain T-loop Regulates CHK2 activity. J Biol Chem 2010; 285:33348-33357. [PMID: 20713355 PMCID: PMC2963420 DOI: 10.1074/jbc.m110.149609] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Chk2 is a critical regulator of the cellular DNA damage repair response. Activation of Chk2 in response to IR-induced damage is initiated by phosphorylation of the Chk2 SQ/TQ cluster domain at Ser19, Ser33, Ser35, and Thr68. This precedes autophosphorylation of Thr383/Thr387 in the T-loop region of the kinase domain an event that is a prerequisite for efficient kinase activity. We conducted an in-depth analysis of phosphorylation within the T-loop region (residues 366–406). We report four novel phosphorylation sites at Ser372, Thr378, Thr389, and Tyr390. Substitution mutation Y390F was defective for kinase function. The substitution mutation T378A ablated the IR induction of kinase activity. Interestingly, the substitution mutation T389A demonstrated a 6-fold increase in kinase activity when compared with wild-type Chk2. In addition, phosphorylation at Thr389 was a prerequisite to phosphorylation at Thr387 but not at Thr383. Quantitative mass spectrometry analysis revealed IR-induced phosphorylation and subcellular distribution of Chk2 phosphorylated species. We observed IR-induced increase in phosphorylation at Ser379, Thr389, and Thr383/Thr389. Phosphorylation at Tyr390 was dramatically reduced following IR. Exposure to IR was also associated with changes in the ratio of chromatin/nuclear localization. IR-induced increase in chromatin localization was associated with phosphorylation at Thr372, Thr379, Thr383, Thr389, Thr383/Thr387, and Thr383/Thr389. Chk2 hyper-phosphorylated species at Thr383/Thr387/Thr389 and Thr383/Thr387/Thr389/Tyr390 relocalized from almost exclusively chromatin to predominately nuclear expression, suggesting a role for phosphorylation in regulation of chromatin targeting and egress. The differential impact of T-loop phosphorylation on Chk2 ubiquitylation suggests a co-dependence of these modifications. The results demonstrate that a complex interdependent network of phosphorylation events within the T-loop exchange region regulates dimerization/autophosphorylation, kinase activation, and chromatin targeting/egress of Chk2.
Collapse
Affiliation(s)
- Xin Guo
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Michael D Ward
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Jessica B Tiedebohl
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Yvonne M Oden
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Julius O Nyalwidhe
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - O John Semmes
- From the Department of Microbiology and Molecular Cell Biology, Cancer Biology and Infectious Disease Research Center, Eastern Virginia Medical School, Norfolk, Virginia 23507.
| |
Collapse
|
33
|
Jeang KT, Giam CZ, Majone F, Aboud M. HTLV-1 Tax: Linking transformation, DNA damage and apoptotic T-cell death. J Biol Chem 2010; 279:31991-4. [PMID: 15090550 DOI: 10.1074/jbc.r400009200] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The human T-cell leukemia virus type I (HTLV-1) is the causative agent of adult T-cell leukemia (ATL), an aggressive CD4-positive T-cell neoplasia. The HTLV-1 proto-oncogene Tax, a potent transcriptional activator of cellular and viral genes, is thought to play a pivotal role in the transforming properties of the virus by deregulating intracellular signaling pathways. During the course of HTLV-1 infection, the dysregulation of cell-cycle checkpoints and the suppression of DNA damage repair is tightly linked to the activity of the viral oncoprotein Tax. Tax activity is associated with production of reactive oxygen intermediates (ROS), chromosomal instability and DNA damage, apoptotic cell death and cellular transformation. Changes in the intracellular redox status induced by Tax promote DNA damage. Tax-mediated DNA damage is believed to be essential in initiating the transformation process by subjecting infected T cells to genetic changes that eventually promote the neoplastic state. Apoptosis and immune surveillance would then exert the necessary selection pressure for eliminating the majority of virally infected cells, while escape variants acquiring a mutator phenotype would constitute a subpopulation of genetically altered cells prone to neoplasia. While the potency of Tax-activity seems to be a determining factor for the observed effects, the cooperation of Tax with other viral proteins determines the fate and progression of HTLV-1-infected cells through DNA damage, apoptosis, survival and transformation.
Collapse
Affiliation(s)
- Kuan-Teh Jeang
- Laboratory of Molecular Microbiology, Nattional Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | |
Collapse
|
34
|
Chlichlia K, Khazaie K. HTLV-1 Tax: Linking transformation, DNA damage and apoptotic T-cell death. Chem Biol Interact 2010; 188:359-65. [PMID: 20558150 DOI: 10.1016/j.cbi.2010.06.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 06/01/2010] [Accepted: 06/06/2010] [Indexed: 11/20/2022]
Abstract
The human T-cell leukemia virus type I (HTLV-1) is the causative agent of adult T-cell leukemia (ATL), an aggressive CD4-positive T-cell neoplasia. The HTLV-1 proto-oncogene Tax, a potent transcriptional activator of cellular and viral genes, is thought to play a pivotal role in the transforming properties of the virus by deregulating intracellular signaling pathways. During the course of HTLV-1 infection, the dysregulation of cell-cycle checkpoints and the suppression of DNA damage repair is tightly linked to the activity of the viral oncoprotein Tax. Tax activity is associated with production of reactive oxygen intermediates (ROS), chromosomal instability and DNA damage, apoptotic cell death and cellular transformation. Changes in the intracellular redox status induced by Tax promote DNA damage. Tax-mediated DNA damage is believed to be essential in initiating the transformation process by subjecting infected T cells to genetic changes that eventually promote the neoplastic state. Apoptosis and immune surveillance would then exert the necessary selection pressure for eliminating the majority of virally infected cells, while escape variants acquiring a mutator phenotype would constitute a subpopulation of genetically altered cells prone to neoplasia. While the potency of Tax-activity seems to be a determining factor for the observed effects, the cooperation of Tax with other viral proteins determines the fate and progression of HTLV-1-infected cells through DNA damage, apoptosis, survival and transformation.
Collapse
Affiliation(s)
- Katerina Chlichlia
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | | |
Collapse
|
35
|
Zane L, Sibon D, Jeannin L, Zandecki M, Delfau-Larue MH, Gessain A, Gout O, Pinatel C, Lançon A, Mortreux F, Wattel E. Tax gene expression and cell cycling but not cell death are selected during HTLV-1 infection in vivo. Retrovirology 2010; 7:17. [PMID: 20222966 PMCID: PMC2846874 DOI: 10.1186/1742-4690-7-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 03/11/2010] [Indexed: 01/18/2023] Open
Abstract
Background Adult T cell leukemia results from the malignant transformation of a CD4+ lymphoid clone carrying an integrated HTLV-1 provirus that has undergone several oncogenic events over a 30-60 year period of persistent clonal expansion. Both CD4+ and CD8+ lymphocytes are infected in vivo; their expansion relies on CD4+ cell cycling and on the prevention of CD8+ cell death. Cloned infected CD4+ but not CD8+ T cells from patients without malignancy also add up nuclear and mitotic defects typical of genetic instability related to theexpression of the virus-encoded oncogene tax. HTLV-1 expression is cancer-prone in vitro, but in vivo numerous selection forces act to maintain T cell homeostasis and are possibly involved in clonal selection. Results Here we demonstrate that the HTLV-1 associated CD4+ preleukemic phenotype and the specific patterns of CD4+ and CD8+ clonal expansion are in vivo selected processes. By comparing the effects of recent (1 month) experimental infections performed in vitro and those observed in cloned T cells from patients infected for >6-26 years, we found that in chronically HTLV-1 infected individuals, HTLV-1 positive clones are selected for tax expression. In vivo, infected CD4+ cells are positively selected for cell cycling whereas infected CD8+ cells and uninfected CD4+ cells are negatively selected for the same processes. In contrast, the known HTLV-1-dependent prevention of CD8+ T cell death pertains to both in vivo and in vitro infected cells. Conclusions Therefore, virus-cell interactions alone are not sufficient to initiate early leukemogenesis in vivo.
Collapse
Affiliation(s)
- Linda Zane
- CNRS UMR5239, Université de Lyon, Oncovirologie et Biothérapies, Centre Léon Bérard, 69008 Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Human tumor viruses are associated with a variety of human malignancies, and it is estimated that 15% of all human cancers have a viral etiology. An abnormality in chromosomal ploidy or aneuploidy is a hallmark of cancers. In normal cells, euploidy is governed by several factors including an intact spindle assembly checkpoint, accurate centrosome duplication, and proper cytokinesis. Viral oncoproteins are suggested to perturb the cellular machineries for chromosomal segregation creating aneuploidy which can lead to the malignant transformation of infected cells. Here, we review in brief some of the mechanisms used by viruses that can cause cellular aneuploidy.
Collapse
Affiliation(s)
- Junichiro Yasunaga
- Molecular Virology Section, Laboratory of Molecular Microbiology, The National Institute of Allergy and Infectious Diseases/NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | |
Collapse
|
37
|
Abstract
Recognition and repair of DNA damage is critical for maintaining genomic integrity and suppressing tumorigenesis. In eukaryotic cells, the sensing and repair of DNA damage are coordinated with cell cycle progression and checkpoints, in order to prevent the propagation of damaged DNA. The carefully maintained cellular response to DNA damage is challenged by viruses, which produce a large amount of exogenous DNA during infection. Viruses also express proteins that perturb cellular DNA repair and cell cycle pathways, promoting tumorigenesis in their quest for cellular domination. This review presents an overview of strategies employed by viruses to manipulate DNA damage responses and cell cycle checkpoints as they commandeer the cell to maximize their own viral replication. Studies of viruses have identified key cellular regulators and revealed insights into molecular mechanisms governing DNA repair, cell cycle checkpoints, and transformation.
Collapse
Affiliation(s)
- Mira S. Chaurushiya
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Graduate Program, Division of Biology, University of California, San Diego, CA 92093, USA
| | - Matthew D. Weitzman
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
38
|
Zhou PK, Sun Y, An J. Interaction between viral proteins and hosts and its disturbance in the cellular responses to ionising radiation. Int J Radiat Biol 2009; 85:587-97. [DOI: 10.1080/09553000902954512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
39
|
Lodewick J, Lamsoul I, Polania A, Lebrun S, Burny A, Ratner L, Bex F. Acetylation of the human T-cell leukemia virus type 1 Tax oncoprotein by p300 promotes activation of the NF-kappaB pathway. Virology 2009; 386:68-78. [PMID: 19200568 PMCID: PMC2834172 DOI: 10.1016/j.virol.2008.12.043] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 10/17/2008] [Accepted: 12/31/2008] [Indexed: 01/28/2023]
Abstract
The oncogenic potential of the HTLV-1 Tax protein involves activation of the NF-kappaB pathway, which depends on Tax phosphorylation, ubiquitination and sumoylation. We demonstrate that the nuclei of Tax-expressing cells, including HTLV-1 transformed T-lymphocytes, contain a pool of Tax molecules acetylated on lysine residue at amino acid position 346 by the transcriptional coactivator p300. Phosphorylation of Tax on serine residues 300/301 was a prerequisite for Tax localization in the nucleus and correlated with its subsequent acetylation by p300, whereas sumoylation, resulting in the formation of Tax nuclear bodies in which p300 was recruited, favored Tax acetylation. Overexpression of p300 markedly increased Tax acetylation and the ability of a wild type HTLV-1 provirus, but not of a mutant provirus carrying an acetylation deficient Tax gene, to activate gene expression from an integrated NF-kappaB-controlled promoter. Thus, Tax acetylation favors NF-kappaB activation and might play an important role in HTLV-1-induced cell transformation.
Collapse
Affiliation(s)
- Julie Lodewick
- Institute for Microbiological Research J-M Wiame and Laboratory of Microbiology, Université Libre de Bruxelles, 1, Avenue Emile Gryson, B-1070 Brussels, Belgium
| | - Isabelle Lamsoul
- Institute for Microbiological Research J-M Wiame and Laboratory of Microbiology, Université Libre de Bruxelles, 1, Avenue Emile Gryson, B-1070 Brussels, Belgium
| | - Angela Polania
- Institute for Microbiological Research J-M Wiame and Laboratory of Microbiology, Université Libre de Bruxelles, 1, Avenue Emile Gryson, B-1070 Brussels, Belgium
| | - Sylvie Lebrun
- Institute for Microbiological Research J-M Wiame and Laboratory of Microbiology, Université Libre de Bruxelles, 1, Avenue Emile Gryson, B-1070 Brussels, Belgium
| | - Arsène Burny
- Faculté des Sciences Agronomiques de Gembloux, Gembloux, Belgium
| | - Lee Ratner
- Division of Molecular Oncology, Washington University School of Medicine, St Louis, USA
| | - Françoise Bex
- Institute for Microbiological Research J-M Wiame and Laboratory of Microbiology, Université Libre de Bruxelles, 1, Avenue Emile Gryson, B-1070 Brussels, Belgium
| |
Collapse
|
40
|
Dimerization and a novel Tax speckled structure localization signal are required for Tax nuclear localization. J Virol 2009; 83:5339-52. [PMID: 19321601 DOI: 10.1128/jvi.00232-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human T-cell leukemia virus type 1 oncoprotein Tax has pleiotropic activities, a subset of which likely leads to immortalization of T cells. Tax is expressed and known to function in both the cell nucleus and the cytoplasm. Tax has defined nuclear localization (NLS) and nuclear export signals that enable shuttling between the two compartments. In this study, we identified a novel region in Tax that targets the protein to discrete nuclear foci that we have previously termed Tax speckled structures (TSS). We demonstrated that the identified region is both necessary and sufficient for directing proteins to TSS. This novel TSS localization signal (TSLS), spanning amino acids 50 to 75, is separable from and adjacent to the NLS of Tax. Coexpression of a Tax NLS mutant and a Tax TSLS mutant rescued the nuclear entry and subnuclear TSS targeting of both proteins, demonstrating that these signals are independent domains. Our analysis also revealed that Tax proteins deficient for dimerization fail to localize to the nucleus. Consequently, when we restored dimerization via induction of a heterologous "dimerizer" domain, nuclear localization was rescued. Thus, we defined additional domains in Tax specific for nuclear localization and subnuclear targeting. Our results reveal a more complex network for regulation of Tax subcellular localization and subsequent function.
Collapse
|
41
|
Durkin SS, Guo X, Fryrear KA, Mihaylova VT, Gupta SK, Belgnaoui SM, Haoudi A, Kupfer GM, Semmes OJ. HTLV-1 Tax oncoprotein subverts the cellular DNA damage response via binding to DNA-dependent protein kinase. J Biol Chem 2008; 283:36311-20. [PMID: 18957425 PMCID: PMC2605996 DOI: 10.1074/jbc.m804931200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Human T-cell leukemia virus type-1 is the causative agent for adult T-cell leukemia. Previous research has established that the viral oncoprotein Tax mediates the transformation process by impairing cell cycle control and cellular response to DNA damage. We showed previously that Tax sequesters huChk2 within chromatin and impairs the response to ionizing radiation. Here we demonstrate that DNA-dependent protein kinase (DNA-PK) is a member of the Tax.Chk2 nuclear complex. The catalytic subunit, DNA-PKcs, and the regulatory subunit, Ku70, were present. Tax-containing nuclear extracts showed increased DNA-PK activity, and specific inhibition of DNA-PK prevented Tax-induced activation of Chk2 kinase activity. Expression of Tax induced foci formation and phosphorylation of H2AX. However, Tax-induced constitutive signaling of the DNA-PK pathway impaired cellular response to new damage, as reflected in suppression of ionizing radiation-induced DNA-PK phosphorylation and gammaH2AX stabilization. Tax co-localized with phospho-DNA-PK into nuclear speckles and a nuclear excluded Tax mutant sequestered endogenous phospho-DNA-PK into the cytoplasm, suggesting that Tax interaction with DNA-PK is an initiating event. We also describe a novel interaction between DNA-PK and Chk2 that requires Tax. We propose that Tax binds to and stabilizes a protein complex with DNA-PK and Chk2, resulting in a saturation of DNA-PK-mediated damage repair response.
Collapse
Affiliation(s)
- Sarah S Durkin
- Department of Microbiology and Molecular Cell Biology, Center for Biomedical Proteomics, Eastern Virginia Medical School, Norfolk, Virginia 23507, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Boxus M, Twizere JC, Legros S, Dewulf JF, Kettmann R, Willems L. The HTLV-1 Tax interactome. Retrovirology 2008; 5:76. [PMID: 18702816 PMCID: PMC2533353 DOI: 10.1186/1742-4690-5-76] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Accepted: 08/14/2008] [Indexed: 12/22/2022] Open
Abstract
The Tax1 oncoprotein encoded by Human T-lymphotropic virus type I is a major determinant of viral persistence and pathogenesis. Tax1 affects a wide variety of cellular signalling pathways leading to transcriptional activation, proliferation and ultimately transformation. To carry out these functions, Tax1 interacts with and modulates activity of a number of cellular proteins. In this review, we summarize the present knowledge of the Tax1 interactome and propose a rationale for the broad range of cellular proteins identified so far.
Collapse
Affiliation(s)
- Mathieu Boxus
- University Academia Wallonie-Europe, Molecular and Cellular Biology at FUSAGx, Gembloux, Belgium.
| | | | | | | | | | | |
Collapse
|
43
|
Human T-cell leukemia virus type 1 infection leads to arrest in the G1 phase of the cell cycle. J Virol 2008; 82:8442-55. [PMID: 18596104 DOI: 10.1128/jvi.00091-08] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Infection by the human T-cell leukemia virus type 1 (HTLV-1) is thought to cause dysregulated T-cell proliferation, which in turn leads to adult T-cell leukemia/lymphoma. Early cellular changes after HTLV-1 infection have been difficult to study due to the poorly infectious nature of HTLV-1 and the need for cell-to-cell contact for HTLV-1 transmission. Using a series of reporter systems, we show that HeLa cells cease proliferation within one or two division cycles after infection by HTLV-1 or transduction of the HTLV-1 tax gene. HTLV-1-infected HeLa cells, like their tax-transduced counterparts, expressed high levels of p21(CIP1/WAF1) and p27(KIP1), developed mitotic abnormalities, and became arrested in G(1) in senescence. In contrast, cells of a human osteosarcoma lineage (HOS) continued to divide after HTLV-1 infection or Tax expression, albeit at a reduced growth rate and with mitotic aberrations. Unique to HOS cells is the dramatic reduction of p21(CIP1/WAF1) and p27(KIP1) expression, which is in part associated with the constitutive activation of the phosphatidylinositol-3-kinase (PI3K)-protein kinase B (Akt) pathway. The loss of p21(CIP1/WAF1) and p27(KIP1) in HOS cells apparently allows HTLV-1- and Tax-induced G(1) arrest to be bypassed. Finally, HTLV-1 infection and Tax expression also cause human SupT1 T cells to arrest in the G(1) phase of the cell cycle. These results suggest that productive HTLV-1 infection ordinarily leads to Tax-mediated G(1) arrest. However, T cells containing somatic mutations that inactivate p21(CIP1/WAF1) and p27(KIP1) may continue to proliferate after HTLV-1 infection and Tax expression. These infected cells can expand clonally, accumulate additional chromosomal abnormalities, and progress to cancer.
Collapse
|
44
|
Human T-cell leukemia virus type 1 tax attenuates the ATM-mediated cellular DNA damage response. J Virol 2008; 82:6952-61. [PMID: 18434398 DOI: 10.1128/jvi.02331-07] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Genomic instability, a hallmark of leukemic cells, is associated with malfunctioning cellular responses to DNA damage caused by defective cell cycle checkpoints and/or DNA repair. Adult T-cell leukemia, which can result from infection with human T-cell leukemia virus type 1 (HTLV-1), is associated with extensive genomic instability that has been attributed to the viral oncoprotein Tax. How Tax influences cellular responses to DNA damage to mediate genomic instability, however, remains unclear. Therefore, we investigated the effect of Tax on cellular pathways involved in recognition and repair of DNA double-strand breaks. Premature attenuation of ATM kinase activity and reduced association of MDC1 with repair foci were observed in Tax-expressing cells. Following ionizing radiation-induced S-phase checkpoint activation, Tax-expressing cells progressed more rapidly than non-Tax-expressing cells toward DNA replication. These results demonstrate that Tax expression may allow premature DNA replication in the presence of genomic lesions. Attempts to replicate in the presence of these lesions would result in gradual accumulation of mutations, leading to genome instability and cellular transformation.
Collapse
|
45
|
Schavinsky-Khrapunsky Y, Priel E, Aboud M. Dose-dependent dual effect of HTLV-1 tax oncoprotein on p53-dependent nucleotide excision repair in human T-cells. Int J Cancer 2008; 122:305-16. [PMID: 17918160 DOI: 10.1002/ijc.23091] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this study we investigated the effect of Tax on nucleotide excision repair (NER) in human T-cell lines by using the host cell repair analysis of UVC-irradiated reporter plasmid. This analysis revealed a p53-dependent NER activity in wild type (w.t.) p53-containing T-cells and p53-independent NER in w.t. p53-lacking T-cells. Notably, in the w.t. p53-containing cells Tax exerted a dose-dependent dual effect on NER. While low Tax doses markedly stimulated this repair, high Tax doses strongly reduced it. Further experiments demonstrated that the low Tax doses enhanced, in these cells, the level and the transcriptional function of their w.t. p53 protein. On the other hand, although the high Tax doses further increased the level of p53, they functionally inactivated its accumulating molecules. Both of these Tax effects on p53 proved to be mediated by Tax-induced NF-kappaB-related mechanisms. Together, these data suggest that by NF-kappaB activation Tax elevates the level of the cellular w.t. p53. However, while at low Tax doses the elevating w.t. p53 molecules are functionally active and capable of stimulating NER, intensifying further the NF-kappaB activation by the high Tax doses concomitantly evokes certain mechanism(s) which functionally inactivates the accumulating p53 protein. In contrast to this dual effect on the p53-dependent NER, Tax displayed only an inhibitory effect on the p53-independent NER by its high doses, whereas its low doses had no effect on this repair. The mechanisms of the NF-kappaB-associated effects on the level and function of the cellular w.t.p53 and of the p53-independent NER noted in our experimental systems are further investigated in our laboratory.
Collapse
Affiliation(s)
- Yana Schavinsky-Khrapunsky
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences and Cancer Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | | | | |
Collapse
|
46
|
Peloponese JM, Kinjo T, Jeang KT. Human T-cell leukemia virus type 1 Tax and cellular transformation. Int J Hematol 2007; 86:101-6. [PMID: 17875521 DOI: 10.1532/ijh97.07087] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Infection of T-cells by human T-cell leukemia virus type 1 (HTLV-1) causes a lymphoproliferative malignancy known as adult T-cell leukemia (ATL). ATL is characterized by abnormal lymphocytes, called flower cells, which have cleaved and convoluted nuclei. Tax, encoded by the HTLV-1 pX region, is a critical nonstructural protein that plays a central role in leukemogenesis; however, the mechanisms of HTLV-1 oncogenesis have not been clarified fully. In this review, we summarize current thinking on how Tax may affect ATL leukemogenesis.
Collapse
Affiliation(s)
- Jean-Marie Peloponese
- Molecular Virology Section, Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-0460, USA
| | | | | |
Collapse
|
47
|
Gupta SK, Guo X, Durkin SS, Fryrear KF, Ward MD, Semmes OJ. Human T-cell Leukemia Virus Type 1 Tax Oncoprotein Prevents DNA Damage-induced Chromatin Egress of Hyperphosphorylated Chk2. J Biol Chem 2007; 282:29431-40. [PMID: 17698850 DOI: 10.1074/jbc.m704110200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
De novo expression of human T-cell leukemia virus type 1 Tax results in cellular genomic instability. We demonstrated previously that Tax associates with the cell cycle check point regulator Chk2 and proposed that the inappropriate activation of Chk2 provides a model for Tax-induced loss of genetic integrity (Haoudi, A., Daniels, R. C., Wong, E., Kupfer, G., and Semmes, O. J. (2003) J. Biol. Chem. 278, 37736-37744). Here we provide an explanation for how Tax induces some Chk2 activities but represses others. We show that Tax interaction with Chk2 generates two activation signals in Chk2, oligomerization and autophosphorylation. However, egress of Chk2 from chromatin, normally observed in response to ionizing radiation, was repressed in Tax-expressing cells. Analysis of chromatin-bound Chk2 from Tax-expressing cells revealed phosphorylation at Thr(378), Ser(379), Thr(383), Thr(387), and Thr(389). In contrast, chromatin-bound Chk2 in the absence of Tax was phosphorylated at Thr(383) and Thr(387) in response to ionizing radiation. We further establish that Tax binds to the kinase domain of Chk2. Confocal microscopy revealed a redistribution of Chk2 to colocalize with Tax in Tax speckled structures, which we have shown previously to coincide with interchromatin granules. We propose that Tax binding via the Chk2 kinase domain sequesters phosphorylated Chk2 within chromatin, thus hindering chromatin egress and appropriate response to DNA damage.
Collapse
Affiliation(s)
- Saurabh K Gupta
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507, USA
| | | | | | | | | | | |
Collapse
|
48
|
Datta A, Nicot C. Telomere attrition induces a DNA double-strand break damage signal that reactivates p53 transcription in HTLV-I leukemic cells. Oncogene 2007; 27:1135-41. [PMID: 17704807 DOI: 10.1038/sj.onc.1210718] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Persistent inhibition of telomerase induces a severe telomere shortening in human T-cell leukemia virus type-1-infected cells which signals a DNA double-strand break damage response, formation of telomere dysfunction-induced foci and activates the ATM pathway. In turn, activation of ATM and its downstream effectors led to an increased phosphorylation and acetylation on specific residues of p53 known to be involved in transcriptional activation. Disruption of Mdm2-p53 complexes coupled with increased proteasomal degradation of MDMX further enhanced reactivation of p53 transcription, ultimately leading to senescence of tumor cells. Induction of senescence in these T-cells was associated with an increased expression of p21, p16 and activation of GSK3beta. Our results support the cancer-aging model and demonstrate that the halt of aging in cancer cells can be reversed through reactivation of p53.
Collapse
Affiliation(s)
- A Datta
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | |
Collapse
|
49
|
Abstract
The cAMP response element-binding protein (CREB) is a stimulus-induced transcription factor that responds rapidly to phosphorylation and/or coactivator activation. Regulated activation of CREB has a significant impact on cellular growth, proliferation and survival. To overturn the cellular control of these processes, tumor cells have developed various mechanisms to achieve constitutive activation of CREB, including gene amplification, chromosome translocation, interaction with viral oncoproteins, and inactivation of tumor suppressor genes. These mechanisms converge on the phosphorylation of CREB and/or the activation of transducer of regulated CREB activity (TORC) coactivators to effect uncontrolled proliferation of cells. This minireview summarizes the different lines of existing evidence that support a direct role of CREB in oncogenesis.
Collapse
Affiliation(s)
- Yeung-Tung Siu
- Department of Biochemistry, The University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
50
|
Abstract
Par 6 acts as a scaffold protein to facilitate atypical protein kinase C-mediated phosphorylation of cytoplasmic protein complexes, leading to epithelial and neuronal cell polarization. In addition to its location in the cytoplasm, Par 6 is localized to the nucleus. However, its organization and potential functions in the nucleus have not been examined. Using an affinity-purified Par 6 antibody and a chimera of Par 6 and green fluorescent protein, we show that Par 6 localizes precisely to nuclear speckles, but not to other nuclear structures, and displays characteristics of speckle proteins. We show that Par 6 colocalizes in the nucleus with Tax, a transcriptional activator of the human T-cell leukemia virus type 1 long terminal repeat, but multiple lines of evidence show that Par 6 is not directly involved in known functions of speckle proteins, including general transcription, splicing, or mRNA transport. Significantly, however, the structure of nuclear speckles is lost when Par 6 levels are reduced by Par 6-specific small interfering RNA. Therefore, we hypothesize that Par 6 in the nucleus acts as a scaffolding protein in nuclear speckle complexes, similar to its role in the cytoplasm.
Collapse
Affiliation(s)
- Erin G Cline
- Department of Biological Sciences, The James A Clark Center, Stanford University, Stanford, CA 94305-5430, USA
| | | |
Collapse
|