1
|
Dubey AK, Singh A, Prakash S, Kumar M, Singh AK. Race to arsenal COVID-19 therapeutics: Current alarming status and future directions. Chem Biol Interact 2020; 332:109298. [PMID: 33121920 PMCID: PMC7588316 DOI: 10.1016/j.cbi.2020.109298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 01/08/2023]
Abstract
The on-going pandemic of COVID-19 wreaked by a viral infection of SARS-CoV-2, has generated a catastrophic plight across the globe. Interestingly, one of the hallmarks of COVID-19 is the so-called 'cytokine storm' due to attack of SARS-Cov-2 in the lungs. Considering, mesenchymal stem cells (MSCs) therapy could contribute against SARS-CoV-2 viruses attack because of their immune modulatory and anti-inflammatory ability linked to their stemness, to the arsenal of treatments for COVID-19. Another novel therapeutic strategies include the blockade of rampant generation of pro-inflammatory mediators like acute respiratory distress syndrome (ARDS), degradation of viral protein capsids by PROTACs, composed of Ubiquitin-proteasome framework, and ubiquitination-independent pathway directing the SARS-CoV-2 nucleocapsid protein (nCoV N) and proteasome activator (PA28γ), etc. This review is consequently an endeavour to highlight the several aspects of COVID-19 with incorporation of important treatment strategies discovered to date and putting the real effort on the future directions to put them into the perspective.
Collapse
Affiliation(s)
- Ankit Kumar Dubey
- Department of Biotechnology, Indian Institute of Technology Madras, Tamil Nadu, 600036, India
| | - Aakansha Singh
- CSIR-Central Drug Research Institute, Lucknow, 226014, India
| | - Shardendu Prakash
- Department of Pharmacy, Sardar Patel College of Pharmacy, Gorakhpur, 273013, India
| | - Manoj Kumar
- Department of Microbiology, SGPGIMS, Lucknow, 226014, India
| | - Ashok K Singh
- Pennsylvania State University, Penn State College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
2
|
Zhang H, Tu J, Cao C, Yang T, Gao L. Proteasome activator PA28γ-dependent degradation of coronavirus disease (COVID-19) nucleocapsid protein. Biochem Biophys Res Commun 2020; 529:251-256. [PMID: 32703419 PMCID: PMC7296323 DOI: 10.1016/j.bbrc.2020.06.058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
Abstract
The nucleocapsid protein is significant in the formation of viral RNA of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), accounting for the largest proportion of viral structural proteins. Here, we report for the first time that the 11S proteasomal activator PA28γ regulates the intracellular abundance of the SARS-CoV-2 N protein (nCoV N). Furthermore, we have identified proteasome activator PA28γ as a nCoV N binding protein by co-immunoprecipitation assay. As a result of their interaction, nCoV N could be degraded by PA28γ-20S in vitro degradation assay. This was also demonstrated by blocking de novo protein synthesis with cycloheximide. The stability of nCoV N in PA28γ-knockout cells was greater than in PA28γ-wildtype cells. Notably, immunofluorescence staining revealed that knockout of the PA28γ gene in cells led to the transport of nCoV N from the nucleus to the cytoplasm. Overexpression of PA28γ enhanced proteolysis of nCoV N compared to that in PA28γ-N151Y cells containing a dominant-negative PA28γ mutation, which reduced this process. These results suggest that PA28γ binding is important in regulating 20S proteasome activity, which in turn regulates levels of the critical nCoV N nucleocapsid protein of SARS-CoV-2, furthering our understanding of the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Haiyang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Jialu Tu
- Shanghai Key Laboratory of Regulatory Biology, Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Chulei Cao
- Shanghai Key Laboratory of Regulatory Biology, Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Ting Yang
- Shanghai Key Laboratory of Regulatory Biology, Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Liangcai Gao
- Shanghai Key Laboratory of Regulatory Biology, Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China.
| |
Collapse
|
3
|
Harrod R. Silencers of HTLV-1 and HTLV-2: the pX-encoded latency-maintenance factors. Retrovirology 2019; 16:25. [PMID: 31492165 PMCID: PMC6731619 DOI: 10.1186/s12977-019-0487-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/30/2019] [Indexed: 11/10/2022] Open
Abstract
Of the members of the primate T cell lymphotropic virus (PTLV) family, only the human T-cell leukemia virus type-1 (HTLV-1) causes disease in humans—as the etiological agent of adult T-cell leukemia/lymphoma (ATLL), HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), and other auto-inflammatory disorders. Despite having significant genomic organizational and structural similarities, the closely related human T-cell lymphotropic virus type-2 (HTLV-2) is considered apathogenic and has been linked with benign lymphoproliferation and mild neurological symptoms in certain infected patients. The silencing of proviral gene expression and maintenance of latency are central for the establishment of persistent infections in vivo. The conserved pX sequences of HTLV-1 and HTLV-2 encode several ancillary factors which have been shown to negatively regulate proviral gene expression, while simultaneously activating host cellular proliferative and pro-survival pathways. In particular, the ORF-II proteins, HTLV-1 p30II and HTLV-2 p28II, suppress Tax-dependent transactivation from the viral promoter—whereas p30II also inhibits PU.1-mediated inflammatory-signaling, differentially augments the expression of p53-regulated metabolic/pro-survival genes, and induces lymphoproliferation which could promote mitotic proviral replication. The ubiquitinated form of the HTLV-1 p13II protein localizes to nuclear speckles and interferes with recruitment of the p300 coactivator by the viral transactivator Tax. Further, the antisense-encoded HTLV-1 HBZ and HTLV-2 APH-2 proteins and mRNAs negatively regulate Tax-dependent proviral gene expression and activate inflammatory signaling associated with enhanced T-cell lymphoproliferation. This review will summarize our current understanding of the pX latency-maintenance factors of HTLV-1 and HTLV-2 and discuss how these products may contribute to the differences in pathogenicity between the human PTLVs.
Collapse
Affiliation(s)
- Robert Harrod
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX, 75275-0376, USA.
| |
Collapse
|
4
|
Namdev P, Lyngdoh DL, Dar HY, Chaurasiya SK, Srivastava R, Tripathi T, Anupam R. Intrinsically Disordered Human T Lymphotropic Virus Type 1 p30 Protein: Experimental and Computational Evidence. AIDS Res Hum Retroviruses 2019; 35:477-487. [PMID: 30618266 DOI: 10.1089/aid.2018.0196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human T lymphotropic virus type 1 (HTLV-1) causes adult T cell leukemia and lymphoma and other neuroinflammatory diseases. The pX region of HTLV-1 genome encodes an accessory protein p30 that is required for viral persistence and spread in the host. p30 regulates viral gene expression at the transcription level by competing with Tax for p300 binding and at posttranscriptional level by nuclear retention of tax/rex messenger RNA (mRNA). In addition, p30 modulates the host cellular environment by binding to various host proteins such as ATM, REGγ, and PRMT5. However, the low expression levels of p30 has been a major hurdle in studying its structure-function relationship in the context of HTLV-1 pathobiology, which is most likely due to its intrinsically disordered nature. To investigate the unstable nature of p30, flow cytometric analysis of p30-GFP fusion protein expressed in Escherichia coli was conducted and bioinformatics analysis of p30 was performed. The bacterial cells were green fluorescent protein (GFP) positive, indicating that p30-GFP was in the soluble fraction. Induction, particularly at higher temperature, reduced the expression of p30-GFP. Moreover, p30-GFP was detected exclusively in insoluble fraction upon cell lysis, suggesting its unstable and disordered nature. The bioinformatics analysis of p30 protein sequence and amino acid content revealed that p30 has highly disordered regions from amino acids 75-155 and 197-241. Furthermore, p30 has regions for macromolecular interactions that could stabilize it and these regions coincide with the unstable regions. Collectively, the study indicates that HTLV-1 p30 is an intrinsically disordered protein.
Collapse
Affiliation(s)
- Priyanka Namdev
- Department of Biotechnology, Dr. Harisingh Gour University, Sagar, India
| | - Denzelle Lee Lyngdoh
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North Eastern Hill University, Shillong, India
| | - Hamid Y. Dar
- Department of Zoology, Dr. Harisingh Gour University, Sagar, India
| | - Shivendra K. Chaurasiya
- Host-Pathogen Interaction and Signal Transduction Laboratory, Department of Microbiology, Dr. Harisingh Gour University, Sagar, India
| | | | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North Eastern Hill University, Shillong, India
| | - Rajaneesh Anupam
- Department of Biotechnology, Dr. Harisingh Gour University, Sagar, India
| |
Collapse
|
5
|
Hutchison T, Malu A, Yapindi L, Bergeson R, Peck K, Romeo M, Harrod C, Pope J, Smitherman L, Gwinn W, Ratner L, Yates C, Harrod R. The TP53-Induced Glycolysis and Apoptosis Regulator mediates cooperation between HTLV-1 p30 II and the retroviral oncoproteins Tax and HBZ and is highly expressed in an in vivo xenograft model of HTLV-1-induced lymphoma. Virology 2018; 520:39-58. [PMID: 29777913 DOI: 10.1016/j.virol.2018.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/28/2022]
Abstract
The human T-cell leukemia virus type-1 (HTLV-1) is an oncoretrovirus that infects and transforms CD4+ T-cells and causes adult T-cell leukemia/lymphoma (ATLL) -an aggressive lymphoproliferative disease that is highly refractive to most anticancer therapies. The HTLV-1 proviral genome encodes several regulatory products within a conserved 3' nucleotide sequence, known as pX; however, it remains unclear how these factors might cooperate or dynamically interact in virus-infected cells. Here we demonstrate that the HTLV-1 latency-maintenance factor p30II induces the TP53-induced glycolysis and apoptosis regulator (TIGAR) and counters the oxidative stress, mitochondrial damage, and cytotoxicity caused by the viral oncoproteins Tax and HBZ. The p30II protein cooperates with Tax and HBZ and enhances their oncogenic potential in colony transformation/foci-formation assays. Further, we have shown that TIGAR is highly expressed in HTLV-1-induced tumors associated with oncogene dysregulation and increased angiogenesis in an in vivo xenograft model of HTLV-1-induced T-cell lymphoma. These findings provide the first evidence that p30II likely collaborates as an ancillary factor for the major oncoproteins Tax and HBZ during retroviral carcinogenesis.
Collapse
Affiliation(s)
- Tetiana Hutchison
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Aditi Malu
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Laçin Yapindi
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Rachel Bergeson
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Kendra Peck
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Megan Romeo
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Carolyn Harrod
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Jordan Pope
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Louisa Smitherman
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Wesleigh Gwinn
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States
| | - Lee Ratner
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Courtney Yates
- Laboratory Animal Resource Center, Southern Methodist University, Dallas, TX 75275, United States
| | - Robert Harrod
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, 6501 Airline Drive, 334-DLS, Dallas, TX 75275-0376, United States.
| |
Collapse
|
6
|
Mozhgani SH, Zarei-Ghobadi M, Teymoori-Rad M, Mokhtari-Azad T, Mirzaie M, Sheikhi M, Jazayeri SM, Shahbahrami R, Ghourchian H, Jafari M, Rezaee SA, Norouzi M. Human T-lymphotropic virus 1 (HTLV-1) pathogenesis: A systems virology study. J Cell Biochem 2018; 119:3968-3979. [PMID: 29227540 DOI: 10.1002/jcb.26546] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/01/2017] [Indexed: 12/31/2022]
Abstract
The main mechanisms of interaction between Human T-lymphotropic virus type 1 (HTLV-1) and its hosts in the manifestation of the related disease including HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP) and Adult T-cell leukemia/lymphoma (ATLL) are yet to be determined. It is pivotal to find out the changes in the genes expression toward an asymptomatic or symptomatic states. To this end, the systems virology analysis was performed. Firstly, the differentially expressed genes (DEGs) were taken pairwise among the four sample sets of Normal, Asymptomatic Carriers (ACs), ATLL, and HAM/TSP. Afterwards, the protein-protein interaction networks were reconstructed utilizing the hub genes. In conclusion, the pathways of cells proliferation and transformation were identified in the ACs state. In addition to immune pathways in ATLL, the inflammation and cancer pathways were discened in both diseases of ATLL and HAM/TSP. The outcomes can specify the genes involved in the pathogenesis and help to design the drugs in the future.
Collapse
Affiliation(s)
- Sayed-Hamidreza Mozhgani
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Zarei-Ghobadi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Majid Teymoori-Rad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mirzaie
- Department of Applied Mathematics, Faculty of Mathematical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Sheikhi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed-Mohammad Jazayeri
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Shahbahrami
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohieddin Jafari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed-Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Norouzi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
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
|
8
|
XU XIAOPING, LIU DONGJUAN, JI NING, LI TAIWEN, LI LONGJIANG, JIANG LU, LI JING, ZHANG PING, ZENG XIN, CHEN QIANMING. A novel transcript variant of proteasome activator 28γ: Identification and function in oral cancer cells. Int J Oncol 2015; 47:188-94. [DOI: 10.3892/ijo.2015.2980] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/04/2015] [Indexed: 11/06/2022] Open
|
9
|
Gruner M, Moncsek A, Rödiger S, Kühnhardt D, Feist E, Stohwasser R. Increased proteasome activator 28 gamma (PA28γ) levels are unspecific but correlate with disease activity in rheumatoid arthritis. BMC Musculoskelet Disord 2014; 15:414. [PMID: 25482151 PMCID: PMC4295294 DOI: 10.1186/1471-2474-15-414] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/28/2014] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND PA28γ (also known as Ki, REG gamma, PMSE3), a member of the ubiquitin-and ATP-independent proteasome activator family 11S, has been proved to show proteasome-dependent and -independent effects on several proteins including tumor suppressor p53, cyclin-dependent kinase inhibitor p21 and steroid receptor co-activator 3 (SCR-3). Interestingly, PA28γ is overexpressed in pathological tissue of various cancers affecting e. g. breast, bowl and thyroids. Furthermore, anti-PA28γ autoantibodies have been linked to several autoimmune disorders. The aim of this study was to develop and evaluate a novel and sensitive PA28γ sandwich ELISA for the quantification of PA28γ serum levels in patients with cancer and autoimmune diseases for diagnostic and prognostic purposes. METHODS PA28γ-specific polyclonal antibodies and recombinant His-tagged PA28γ were purified and used to develop a sandwich ELISA for the detection of circulating PA28γ. With this new assay, PA28γ serum levels of patients with various cancers, rheumatoid arthritis (RA), Sjögren's syndrome (SS), adult-onset Still's disease (AOSD) and different connective-tissue diseases (CTD) were compared with healthy control subjects. Anti-PA28γ autoantibodies were additionally confirmed using a newly developed microbead assay. RESULTS The developed PA28γ sandwich ELISA showed a high specificity with a detection limit of 3 ng/ml. A significant up-regulation of circulating PA28γ was detected in the sera of patients with cancer, RA, SS and CTD. A significant correlation was observed dependent on age as well as anti-PA28γ autoantibody levels with circulating PA28γ protein levels. Furthermore, PA28γ serum levels showed a correlation with disease activity in patients with RA under treatment with the T-cell directed biological compound abatacept according to disease activity score 28 (DAS28) and erythrocyte sedimentation rate (ESR). CONCLUSION The application of PA28γ as a novel biomarker for diagnostic purposes of a specific disease is limited, since elevated levels were observed in different disorders. However, the correlation with disease activity in patients with RA suggests a prognostic value, which needs to be addressed by further studies. Therefore our results show that PA28γ is a useful marker which should be included in studies related to novel treatments, e.g. abatacept.
Collapse
Affiliation(s)
- Melanie Gruner
- />Faculty of Natural Sciences, Brandenburg Technical University Cottbus - Senftenberg, Großenhainer Str. 57, D-01968 Senftenberg, Germany
- />Department of Rheumatology and Clinical Immunology and Autoinflammatory Reference Centre at Charité, Charité-Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| | - Anja Moncsek
- />Faculty of Natural Sciences, Brandenburg Technical University Cottbus - Senftenberg, Großenhainer Str. 57, D-01968 Senftenberg, Germany
- />Department of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| | - Stefan Rödiger
- />Faculty of Natural Sciences, Brandenburg Technical University Cottbus - Senftenberg, Großenhainer Str. 57, D-01968 Senftenberg, Germany
| | - Dagmar Kühnhardt
- />Department of Hematology and Oncology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| | - Eugen Feist
- />Department of Rheumatology and Clinical Immunology and Autoinflammatory Reference Centre at Charité, Charité-Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| | - Ralf Stohwasser
- />Faculty of Natural Sciences, Brandenburg Technical University Cottbus - Senftenberg, Großenhainer Str. 57, D-01968 Senftenberg, Germany
| |
Collapse
|
10
|
Lairmore MD. Animal models of bovine leukemia virus and human T-lymphotrophic virus type-1: insights in transmission and pathogenesis. Annu Rev Anim Biosci 2013; 2:189-208. [PMID: 25384140 DOI: 10.1146/annurev-animal-022513-114117] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bovine leukemia virus (BLV) and human T-lymphotrophic virus type-1 (HTLV-1) are related retroviruses associated with persistent and lifelong infections and a low incidence of lymphomas within their hosts. Both viruses can be spread through contact with bodily fluids containing infected cells, most often from mother to offspring through breast milk. Each of these complex retroviruses contains typical gag, pol, and env genes but also unique, nonstructural proteins encoded from the pX region. These nonstructural genes encode the Tax and Rex regulatory proteins, as well as novel proteins essential for viral spread in vivo. Improvements in the molecular tools to test these viral determinants in cellular and animal models have provided new insights into the pathogenesis of each virus. Comparisons of BLV and HTLV-1 provide insights into mechanisms of spread and tumor formation, as well as potential approaches to therapeutic intervention against the infections.
Collapse
Affiliation(s)
- Michael D Lairmore
- School of Veterinary Medicine, University of California, Davis, California, 95616;
| |
Collapse
|
11
|
Anupam R, Doueiri R, Green PL. The need to accessorize: molecular roles of HTLV-1 p30 and HTLV-2 p28 accessory proteins in the viral life cycle. Front Microbiol 2013; 4:275. [PMID: 24062732 PMCID: PMC3774995 DOI: 10.3389/fmicb.2013.00275] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/24/2013] [Indexed: 11/25/2022] Open
Abstract
Extensive studies of human T-cell leukemia virus (HTLV)-1 and HTLV-2 over the last three decades have provided detailed knowledge on viral transformation, host–viral interactions and pathogenesis. HTLV-1 is the etiological agent of adult T cell leukemia and multiple neurodegenerative and inflammatory diseases while HTLV-2 disease association remains elusive, with few infected individuals displaying neurodegenerative diseases similar to HTLV-1. The HTLV group of oncoretroviruses has a genome that encodes structural and enzymatic proteins Gag, Pro, and Env, regulatory proteins Tax and Rex, and several accessory proteins from the pX region. Of these proteins, HTLV-1 p30 and HTLV-2 p28 are encoded by the open reading frame II of the pX region. Like most other accessory proteins, p30 and p28 are dispensable for in vitro viral replication and transformation but are required for efficient viral replication and persistence in vivo. Both p30 and p28 regulate viral gene expression at the post-transcriptional level whereas p30 can also function at the transcriptional level. Recently, several reports have implicated p30 and p28 in multiple cellular processes, which provide novel insight into HTLV spread and survival and ultimately pathogenesis. In this review we summarize and compare what is known about p30 and p28, highlighting their roles in viral replication and viral pathogenesis.
Collapse
Affiliation(s)
- Rajaneesh Anupam
- Center for Retrovirus Research, The Ohio State University Columbus, OH, USA ; Department of Veterinary Biosciences, The Ohio State University Columbus, OH, USA
| | | | | |
Collapse
|
12
|
Riedel A, Mofolo B, Avota E, Schneider-Schaulies S, Meintjes A, Mulder N, Kneitz S. Accumulation of splice variants and transcripts in response to PI3K inhibition in T cells. PLoS One 2013; 8:e50695. [PMID: 23383294 PMCID: PMC3562341 DOI: 10.1371/journal.pone.0050695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 10/23/2012] [Indexed: 12/17/2022] Open
Abstract
Background Measles virus (MV) causes T cell suppression by interference with phosphatidylinositol-3-kinase (PI3K) activation. We previously found that this interference affected the activity of splice regulatory proteins and a T cell inhibitory protein isoform was produced from an alternatively spliced pre-mRNA. Hypothesis Differentially regulated and alternatively splice variant transcripts accumulating in response to PI3K abrogation in T cells potentially encode proteins involved in T cell silencing. Methods To test this hypothesis at the cellular level, we performed a Human Exon 1.0 ST Array on RNAs isolated from T cells stimulated only or stimulated after PI3K inhibition. We developed a simple algorithm based on a splicing index to detect genes that undergo alternative splicing (AS) or are differentially regulated (RG) upon T cell suppression. Results Applying our algorithm to the data, 9% of the genes were assigned as AS, while only 3% were attributed to RG. Though there are overlaps, AS and RG genes differed with regard to functional regulation, and were found to be enriched in different functional groups. AS genes targeted extracellular matrix (ECM)-receptor interaction and focal adhesion pathways, while RG genes were mainly enriched in cytokine-receptor interaction and Jak-STAT. When combined, AS/RG dependent alterations targeted pathways essential for T cell receptor signaling, cytoskeletal dynamics and cell cycle entry. Conclusions PI3K abrogation interferes with key T cell activation processes through both differential expression and alternative splicing, which together actively contribute to T cell suppression.
Collapse
Affiliation(s)
- Alice Riedel
- Institute for Virology and Immunobiology, University of Wuerzburg, Versbacher, Wuerzburg, Germany
| | - Boitumelo Mofolo
- Computational Biology Group, Department of Clinical Laboratory Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
| | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, Versbacher, Wuerzburg, Germany
| | | | - Ayton Meintjes
- Computational Biology Group, Department of Clinical Laboratory Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
| | - Nicola Mulder
- Computational Biology Group, Department of Clinical Laboratory Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
| | - Susanne Kneitz
- Department of Physiological Chemistry I, Biocenter, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
13
|
Bai XT, Nicot C. Overview on HTLV-1 p12, p8, p30, p13: accomplices in persistent infection and viral pathogenesis. Front Microbiol 2012; 3:400. [PMID: 23248621 PMCID: PMC3518833 DOI: 10.3389/fmicb.2012.00400] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Indexed: 12/29/2022] Open
Abstract
The human T-lymphotropic virus type-1 (HTLV-1) is etiologically linked to adult T cell leukemia/lymphoma and tropical spastic paraparesis/HTLV-1-associated myelopathy. While the role of Tax and Rex in viral replication and pathogenesis has been extensively studied, recent evidence suggests that additional viral proteins are essential for the virus life cycle in vivo. In this review, we will summarize possible molecular mechanisms evoked in the literature to explain how p12, p8, p30, and p13 facilitate persistent viral infection of the host. We will explore several stratagems used by HTLV-1 accessory genes to escape immune surveillance, to establish latency, and to deregulate cell cycle and apoptosis to participate in virus-mediated cellular transformation.
Collapse
Affiliation(s)
- Xue Tao Bai
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center Kansas City, KS, USA
| | | |
Collapse
|
14
|
Abstract
UNLABELLED The establishment of a latent reservoir by human tumor viruses is a vital step in initiating cellular transformation and represents a major shortcoming to current therapeutic strategies and the ability to eradicate virus-infected cells. Human T-cell leukemia virus type 1 (HTLV-1) establishes a lifelong infection and is linked to adult T-cell leukemia lymphoma (ATLL). Here, we demonstrate that HTLV-1 p30 recruits the cellular proteasome activator PA28γ onto the viral tax/rex mRNA to prevent its nuclear export and suppress virus replication. Interaction of p30 with a PA28γ retaining fully functional proteasome activity is required for p30's ability to repress HTLV-1. Consistently, HTLV-1 molecular clones replicate better and produce more virus particles in PA28γ-deficient cells. These results define a unique and novel role for the cellular factor PA28γ in the control of nuclear RNA trafficking and HTLV-1–induced latency. Importantly, knockdown of PA28γ expression in ATLL cells latently infected with HTLV-1 reactivates expression of viral tax/rex RNA and the Tax protein. Because Tax is the most immunogenic viral antigen and triggers strong CTL responses, our results suggest that PA28γ-targeted therapy may reactivate virus expression from latently infected cells and allow their eradication from the host. KEY POINTS PA28γ acts as a co-repressor of HTLV-1 p30 to suppress virus replication and is required for the maintenance of viral latency. HTLV-1 has evolved a unique function mediated by its posttranscriptional repressor p30, which is not found in HTLV-2.
Collapse
|
15
|
Nakano K, Watanabe T. HTLV-1 Rex: the courier of viral messages making use of the host vehicle. Front Microbiol 2012; 3:330. [PMID: 22973269 PMCID: PMC3434621 DOI: 10.3389/fmicb.2012.00330] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 08/24/2012] [Indexed: 01/25/2023] Open
Abstract
The human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus causing an aggressive T-cell malignancy, adult T-cell leukemia (ATL). Although HTLV-1 has a compact RNA genome, it has evolved elaborate mechanisms to maximize its coding potential. The structural proteins Gag, Pro, and Pol are encoded in the unspliced form of viral mRNA, whereas the Env protein is encoded in singly spliced viral mRNA. Regulatory and accessory proteins, such as Tax, Rex, p30II, p12, and p13, are translated only from fully spliced mRNA. For effective viral replication, translation from all forms of HTLV-1 transcripts has to be achieved in concert, although unspliced mRNA are extremely unstable in mammalian cells. It has been well recognized that HTLV-1 Rex enhances the stability of unspliced and singly spliced HTLV-1 mRNA by promoting nuclear export and thereby removing them from the splicing site. Rex specifically binds to the highly structured Rex responsive element (RxRE) located at the 3' end of all HTLV-1 mRNA. Rex then binds to the cellular nuclear exporter, CRM1, via its nuclear export signal domain and the Rex-viral transcript complex is selectively exported from the nucleus to the cytoplasm for effective translation of the viral proteins. Yet, the mechanisms by which Rex inhibits the cellular splicing machinery and utilizes the cellular pathways beneficial to viral survival in the host cell have not been fully explored. Furthermore, physiological impacts of Rex against homeostasis of the host cell via interactions with numerous cellular proteins have been largely left uninvestigated. In this review, we focus on the biological importance of HTLV-1 Rex in the HTLV-1 life cycle by following the historical path in the literature concerning this viral post-transcriptional regulator from its discovery to this day. In addition, for future studies, we discuss recently discovered aspects of HTLV-1 Rex as a post-transcriptional regulator and its use in host cellular pathways.
Collapse
Affiliation(s)
- Kazumi Nakano
- Laboratory of Tumor Cell Biology, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo Tokyo, Japan
| | - Toshiki Watanabe
- Laboratory of Tumor Cell Biology, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo Tokyo, Japan
| |
Collapse
|
16
|
Doueiri R, Anupam R, Kvaratskhelia M, Green KB, Lairmore MD, Green PL. Comparative host protein interactions with HTLV-1 p30 and HTLV-2 p28: insights into difference in pathobiology of human retroviruses. Retrovirology 2012; 9:64. [PMID: 22876852 PMCID: PMC3464894 DOI: 10.1186/1742-4690-9-64] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 07/11/2012] [Indexed: 02/02/2023] Open
Abstract
Background Human T lymphotropic virus type-1 (HTLV-1) and type 2 (HTLV-2) are closely related human retroviruses, but have unique disease associations. HTLV-1 is the causative agent of an aggressive T-cell leukemia known as adult T-cell leukemia (ATL), HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP), and other inflammatory diseases. HTLV-2 infection has not been clearly associated with any disease condition. Although both viruses can transform T cells in vitro, the HTLV-1 provirus is mainly detected in CD4+ T cells whereas HTLV-2 is mainly detected in CD8+ T cells of infected individuals. HTLV-1 and HTLV-2 encode accessory proteins p30 and p28, respectively, which share partial amino acid homology and are required for viral persistence in vivo. The goal of this study was to identify host proteins interacting with p30 and p28 in order to understand their role in pathogenesis. Results Affinity-tag purification coupled with mass spectrometric (MS) analyses revealed 42 and 22 potential interacting cellular partners of p30 and p28, respectively. Of these, only three cellular proteins, protein arginine methyltransferase 5 (PRMT5), hnRNP K and 60 S ribosomal protein L8 were detected in both p30 and p28 fractions. To validate the proteomic results, four interacting proteins were selected for further analyses using immunoblot assays. In full agreement with the MS analysis two cellular proteins REGγ and NEAF-interacting protein 30 (NIP30) selectively interacted with p30 and not with p28; heterogeneous nuclear ribonucleoprotein H1 (hnRNP H1) bound to p28 and not to p30; and PRMT5 interacted with both p30 and p28. Further studies demonstrated that reduced levels of PRMT5 resulted in decreased HTLV-2 viral gene expression whereas the viral gene expression of HTLV-1 was unchanged. Conclusion The comparisons of p30 and p28 host protein interaction proteome showed striking differences with some degree of overlap. PRMT5, one of the host proteins that interacted with both p30 and p28 differentially affected HTLV-1 and HTLV-2 viral gene expression suggesting that PRMT5 is involved at different stages of HTLV-1 and HTLV-2 biology. These findings suggest that distinct host protein interaction profiles of p30 and p28 could, in part, be responsible for differences in HTLV-1 and HTLV-2 pathobiology. This study provides new avenues of investigation into mechanisms of viral infection, tropism and persistence.
Collapse
Affiliation(s)
- Rami Doueiri
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
17
|
Lairmore MD, Haines R, Anupam R. Mechanisms of human T-lymphotropic virus type 1 transmission and disease. Curr Opin Virol 2012; 2:474-81. [PMID: 22819021 DOI: 10.1016/j.coviro.2012.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/26/2012] [Accepted: 06/26/2012] [Indexed: 10/28/2022]
Abstract
Human T-lymphotrophic virus type-1 (HTLV-1) infects approximately 15-20 million people worldwide, with endemic areas in Japan, the Caribbean, and Africa. The virus is spread through contact with bodily fluids containing infected cells most often from mother to child through breast milk or via blood transfusion. After prolonged latency periods, approximately 3-5% of HTLV-1 infected individuals will develop either adult T-cell leukemia/lymphoma, or other lymphocyte-mediated disorders such as HTLV-1-associated myelopathy/tropical spastic paraparesis. The genome of this complex retrovirus contains typical gag, pol, and env genes, but also unique nonstructural proteins encoded from the pX region. These nonstructural genes encode the Tax and Rex regulatory proteins, as well as novel proteins essential for viral spread in vivo such as p30, p12, p13 and the antisense-encoded HTLV-1 basic leucine zipper factor (HBZ). While progress has been made in knowledge of viral determinants of cell transformation and host immune responses, host and viral determinants of HTLV-1 transmission and spread during the early phases of infection are unclear. Improvements in the molecular tools to test these viral determinants in cellular and animal models have provided new insights into the early events of HTLV-1 infection. This review will focus on studies that test HTLV-1 determinants in context to full-length infectious clones of the virus providing insights into the mechanisms of transmission and spread of HTLV-1.
Collapse
Affiliation(s)
- Michael D Lairmore
- University of California-Davis, School of Veterinary Medicine, Davis, CA 95616, United States.
| | | | | |
Collapse
|
18
|
Tripathi LP, Kambara H, Moriishi K, Morita E, Abe T, Mori Y, Chen YA, Matsuura Y, Mizuguchi K. Proteomic analysis of hepatitis C virus (HCV) core protein transfection and host regulator PA28γ knockout in HCV pathogenesis: a network-based study. J Proteome Res 2012; 11:3664-79. [PMID: 22646850 DOI: 10.1021/pr300121a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) causes chronic liver disease worldwide. HCV Core protein (Core) forms the viral capsid and is crucial for HCV pathogenesis and HCV-induced hepatocellular carcinoma, through its interaction with the host factor proteasome activator PA28γ. Here, using BD-PowerBlot high-throughput Western array, we attempt to further investigate HCV pathogenesis by comparing the protein levels in liver samples from Core-transgenic mice with or without the knockout of PA28γ expression (abbreviated PA28γ(-/-)CoreTG and CoreTG, respectively) against the wild-type (WT). The differentially expressed proteins integrated into the human interactome were shown to participate in compact and well-connected cellular networks. Functional analysis of the interaction networks using a newly developed data warehouse system highlighted cellular pathways associated with vesicular transport, immune system, cellular adhesion, and cell growth and death among others that were prominently influenced by Core and PA28γ in HCV infection. Follow-up assays with in vitro HCV cell culture systems validated VTI1A, a vesicular transport associated factor, which was upregulated in CoreTG but not in PA28γ(-/-)CoreTG, as a novel regulator of HCV release but not replication. Our analysis provided novel insights into the Core-PA28γ interplay in HCV pathogenesis and identified potential targets for better anti-HCV therapy and potentially novel biomarkers of HCV infection.
Collapse
Affiliation(s)
- Lokesh P Tripathi
- National Institute of Biomedical Innovation, 7-6-8 Saito Asagi, Ibaraki, Osaka, 567-0085, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The DNA damage response (DDR) has emerged as a critical tumour suppressor pathway responding to cellular DNA replicative stress downstream of aberrant oncogene over-expression. Recent studies have now implicated the DDR as a sensor of oncogenic virus infection. In this review, we discuss the mechanisms by which tumour viruses activate and also suppress the host DDR. The mechanism of tumour virus induction of the DDR is intrinsically linked to the need for these viruses to promote an S-phase environment to replicate their nucleic acid during infection. However, inappropriate expression of viral oncoproteins can also activate the DDR through various mechanisms including replicative stress, direct interaction with DDR components and induction of reactive oxygen species. Given the growth-suppressive consequences of activating the DDR, tumour viruses have also evolved mechanisms to attenuate these pathways. Aberrant expression of viral oncoproteins may therefore promote tumourigenesis through increased somatic mutation and aneuploidy due to DDR inactivation. This review will focus on the interplay between oncogenic viruses and the DDR with respect to cellular checkpoint control and transformation.
Collapse
Affiliation(s)
- P A Nikitin
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University Medical Center, 213 Research Dr., CARL 424, DUMC 3054, Durham, NC 27710, USA
| | | |
Collapse
|
20
|
Harrod R. Inhibiting HDACs in a preclinical model of HTLV-1-induced adult T-cell lymphoma. Leuk Res 2011; 35:1436-7. [DOI: 10.1016/j.leukres.2011.07.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 07/29/2011] [Accepted: 07/31/2011] [Indexed: 01/29/2023]
|
21
|
Martin F, Bangham CRM, Ciminale V, Lairmore MD, Murphy EL, Switzer WM, Mahieux R. Conference highlights of the 15th International Conference on Human Retrovirology: HTLV and related retroviruses, 4-8 June 2011, Leuven, Gembloux, Belgium. Retrovirology 2011; 8:86. [PMID: 22035054 PMCID: PMC3223150 DOI: 10.1186/1742-4690-8-86] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 10/28/2011] [Indexed: 12/27/2022] Open
Abstract
The June 2011 15th International Conference on Human Retrovirology: HTLV and Related Viruses marks approximately 30 years since the discovery of HTLV-1. As anticipated, a large number of abstracts were submitted and presented by scientists, new and old to the field of retrovirology, from all five continents. The aim of this review is to distribute the scientific highlights of the presentations as analysed and represented by experts in specific fields of epidemiology, clinical research, immunology, animal models, molecular and cellular biology, and virology.
Collapse
Affiliation(s)
- Fabiola Martin
- Centre for Immunology and Infection, Department of Biology, Hull and York Medical School, University of York, York, UK
| | - Charles RM Bangham
- Department of Immunology, Wright-Fleming Institute, Imperial College London, London, UK
| | - Vincenzo Ciminale
- Department of Oncology and Surgical Sciences and Istituto Oncologico Veneto-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padova, Italy
| | - Michael D Lairmore
- Department of Veterinary Biosciences; Centre for Retrovirus Research; and Comprehensive Cancer Centre, The Arthur James Cancer Hospital and Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Edward L Murphy
- University of California San Francisco and Blood Systems Research Institute, San Francisco, California, USA
| | - William M Switzer
- Laboratory Branch, Division of HIV/AIDS Prevention, National Centre for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centres for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Renaud Mahieux
- Retroviral Oncogenesis Laboratory, INSERM-U758 Human Virology, 69364 Lyon cedex 07, France
- Ecole Normale Supérieure de Lyon, 69364 Lyon cedex 07, France
| |
Collapse
|
22
|
Lairmore MD, Anupam R, Bowden N, Haines R, Haynes RAH, Ratner L, Green PL. Molecular determinants of human T-lymphotropic virus type 1 transmission and spread. Viruses 2011; 3:1131-65. [PMID: 21994774 PMCID: PMC3185783 DOI: 10.3390/v3071131] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 07/01/2011] [Accepted: 07/02/2011] [Indexed: 01/23/2023] Open
Abstract
Human T-lymphotrophic virus type-1 (HTLV-1) infects approximately 15 to 20 million people worldwide, with endemic areas in Japan, the Caribbean, and Africa. The virus is spread through contact with bodily fluids containing infected cells, most often from mother to child through breast milk or via blood transfusion. After prolonged latency periods, approximately 3 to 5% of HTLV-1 infected individuals will develop either adult T-cell leukemia/lymphoma (ATL), or other lymphocyte-mediated disorders such as HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). The genome of this complex retrovirus contains typical gag, pol, and env genes, but also unique nonstructural proteins encoded from the pX region. These nonstructural genes encode the Tax and Rex regulatory proteins, as well as novel proteins essential for viral spread in vivo such as, p30, p12, p13 and the antisense encoded HBZ. While progress has been made in the understanding of viral determinants of cell transformation and host immune responses, host and viral determinants of HTLV-1 transmission and spread during the early phases of infection are unclear. Improvements in the molecular tools to test these viral determinants in cellular and animal models have provided new insights into the early events of HTLV-1 infection. This review will focus on studies that test HTLV-1 determinants in context to full length infectious clones of the virus providing insights into the mechanisms of transmission and spread of HTLV-1.
Collapse
Affiliation(s)
- Michael D. Lairmore
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; E-Mails: (R.A.); (N.B.); (R.H.); (R.A.H.H.); (P.L.G.)
- Comprehensive Cancer Center, The Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-614-292-9203; Fax: +1-614-292-6473
| | - Rajaneesh Anupam
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; E-Mails: (R.A.); (N.B.); (R.H.); (R.A.H.H.); (P.L.G.)
| | - Nadine Bowden
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; E-Mails: (R.A.); (N.B.); (R.H.); (R.A.H.H.); (P.L.G.)
| | - Robyn Haines
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; E-Mails: (R.A.); (N.B.); (R.H.); (R.A.H.H.); (P.L.G.)
| | - Rashade A. H. Haynes
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; E-Mails: (R.A.); (N.B.); (R.H.); (R.A.H.H.); (P.L.G.)
| | - Lee Ratner
- Department of Medicine, Pathology, and Molecular Microbiology, Division of Biology and Biological Sciences, Washington University School of Medicine, Campus Box 8069, 660 S. Euclid Ave., St. Louis, MO 63110, USA; E-Mail: (L.R.)
| | - Patrick L. Green
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; E-Mails: (R.A.); (N.B.); (R.H.); (R.A.H.H.); (P.L.G.)
- Comprehensive Cancer Center, The Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
23
|
Orf-I and orf-II-encoded proteins in HTLV-1 infection and persistence. Viruses 2011; 3:861-85. [PMID: 21994758 PMCID: PMC3185781 DOI: 10.3390/v3060861] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 05/25/2011] [Accepted: 05/26/2011] [Indexed: 01/10/2023] Open
Abstract
The 3′ end of the human T-cell leukemia/lymphoma virus type-1 (HTLV-1) genome contains four overlapping open reading frames (ORF) that encode regulatory proteins. Here, we review current knowledge of HTLV-1 orf-I and orf-II protein products. Singly spliced mRNA from orf-I encodes p12, which can be proteolytically cleaved to generate p8, while differential splicing of mRNA from orf-II results in production of p13 and p30. These proteins have been demonstrated to modulate transcription, apoptosis, host cell activation and proliferation, virus infectivity and transmission, and host immune responses. Though these proteins are not essential for virus replication in vitro, p8, p12, p13, and p30 have an important role in the establishment and maintenance of HTLV-1 infection in vivo.
Collapse
|