1
|
Gong M, Myster F, Azouz A, Sanchez Sanchez G, Li S, Charloteaux B, Yang B, Nichols J, Lefevre L, Javaux J, Leemans S, Nivelles O, van Campe W, Roels S, Mostin L, van den Berg T, Davison AJ, Gillet L, Connelley T, Vermijlen D, Goriely S, Vanderplasschen A, Dewals BG. Unraveling clonal CD8 T cell expansion and identification of essential factors in γ-herpesvirus-induced lymphomagenesis. Proc Natl Acad Sci U S A 2024; 121:e2404536121. [PMID: 39088396 PMCID: PMC11317613 DOI: 10.1073/pnas.2404536121] [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: 03/04/2024] [Accepted: 07/01/2024] [Indexed: 08/03/2024] Open
Abstract
Alcelaphine gammaherpesvirus 1 (AlHV-1) asymptomatically persists in its natural host, the wildebeest. However, cross-species transmission to cattle results in the induction of an acute and lethal peripheral T cell lymphoma-like disease (PTCL), named malignant catarrhal fever (MCF). Our previous findings demonstrated an essential role for viral genome maintenance in infected CD8+ T lymphocytes but the exact mechanism(s) leading to lymphoproliferation and MCF remained unknown. To decipher how AlHV-1 dysregulates T lymphocytes, we first examined the global phenotypic changes in circulating CD8+ T cells after experimental infection of calves. T cell receptor repertoire together with transcriptomics and epigenomics analyses demonstrated an oligoclonal expansion of infected CD8+ T cells displaying effector and exhaustion gene signatures, including GZMA, GNLY, PD-1, and TOX2 expression. Then, among viral genes expressed in infected CD8+ T cells, we uncovered A10 that encodes a transmembrane signaling protein displaying multiple tyrosine residues, with predicted ITAM and SH3 motifs. Impaired A10 expression did not affect AlHV-1 replication in vitro but rendered AlHV-1 unable to induce MCF. Furthermore, A10 was phosphorylated in T lymphocytes in vitro and affected T cell signaling. Finally, while AlHV-1 mutants expressing mutated forms of A10 devoid of ITAM or SH3 motifs (or both) were able to induce MCF, a recombinant virus expressing a mutated form of A10 unable to phosphorylate its tyrosine residues resulted in the lack of MCF and protected against a wild-type virus challenge. Thus, we could characterize the nature of this γ-herpesvirus-induced PTCL-like disease and identify an essential mechanism explaining its development.
Collapse
Affiliation(s)
- Meijiao Gong
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Françoise Myster
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Abdulkader Azouz
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
- Center for Research in Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
| | - Guillem Sanchez Sanchez
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
- Center for Research in Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, Brussels1050, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, Wavre1300, Belgium
| | - Shifang Li
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Benoit Charloteaux
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), GIGA-Genomics core facility, University of Liège, Liège4000, Belgium
| | - Bin Yang
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Jenna Nichols
- Medical Research Council (MRC)-University of Glasgow Centre for Virus Research, GlasgowG61 1QH, United Kingdom
| | - Lucas Lefevre
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, MidlothianEH25 9RG, United Kingdom
| | - Justine Javaux
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Sylvain Leemans
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Olivier Nivelles
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Willem van Campe
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Machelen 1830, Belgium
| | - Stefan Roels
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Machelen 1830, Belgium
| | - Laurent Mostin
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Machelen 1830, Belgium
| | - Thierry van den Berg
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Machelen 1830, Belgium
| | - Andrew J. Davison
- Medical Research Council (MRC)-University of Glasgow Centre for Virus Research, GlasgowG61 1QH, United Kingdom
| | - Laurent Gillet
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
| | - Timothy Connelley
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, MidlothianEH25 9RG, United Kingdom
| | - David Vermijlen
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
- Center for Research in Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, Brussels1050, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, Wavre1300, Belgium
| | - Stanislas Goriely
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
- Center for Research in Immunology, Université Libre de Bruxelles, Gosselies6041, Belgium
| | - Alain Vanderplasschen
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, Wavre1300, Belgium
| | - Benjamin G. Dewals
- Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine—Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, Liège4000, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, Wavre1300, Belgium
| |
Collapse
|
2
|
Gong M, Myster F, van Campe W, Roels S, Mostin L, van den Berg T, Vanderplasschen A, Dewals BG. Wildebeest-Derived Malignant Catarrhal Fever: A Bovine Peripheral T Cell Lymphoma Caused by Cross-Species Transmission of Alcelaphine Gammaherpesvirus 1. Viruses 2023; 15:v15020526. [PMID: 36851740 PMCID: PMC9968110 DOI: 10.3390/v15020526] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Gammaherpesviruses (γHVs) include viruses that can induce lymphoproliferative diseases and tumors. These viruses can persist in the long term in the absence of any pathological manifestation in their natural host. Alcelaphine gammaherpesvirus 1 (AlHV-1) belongs to the genus Macavirus and asymptomatically infects its natural host, the wildebeest (Connochaetes spp.). However, when transmitted to several susceptible species belonging to the order Artiodactyla, AlHV-1 is responsible for the induction of a lethal lymphoproliferative disease, named wildebeest-derived malignant catarrhal fever (WD-MCF). Understanding the pathogenic mechanisms responsible for the induction of WD-MCF is important to better control the risks of transmission and disease development in susceptible species. The aim of this review is to synthesize the current knowledge on WD-MCF with a particular focus on the mechanisms by which AlHV-1 induces the disease. We discuss the potential mechanisms of pathogenesis from viral entry into the host to the maintenance of viral genomes in infected CD8+ T lymphocytes, and we present current hypotheses to explain how AlHV-1 infection induces a peripheral T cell lymphoma-like disease.
Collapse
Affiliation(s)
- Meijiao Gong
- Laboratory of Immunology-Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Avenue de Cureghem 10, B-4000 Liège, Belgium
- Laboratory of Parasitology, Faculty of Veterinary Medicine, FARAH, ULiège, Avenue de Cureghem 10, B-4000 Liège, Belgium
| | - Françoise Myster
- Laboratory of Immunology-Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Avenue de Cureghem 10, B-4000 Liège, Belgium
| | - Willem van Campe
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Kerklaan 68, B-1830 Machelen, Belgium
| | - Stefan Roels
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Kerklaan 68, B-1830 Machelen, Belgium
| | - Laurent Mostin
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Kerklaan 68, B-1830 Machelen, Belgium
| | - Thierry van den Berg
- Sciensano, Scientific Directorate Infectious Diseases in Animals, Experimental Center Machelen, Kerklaan 68, B-1830 Machelen, Belgium
| | - Alain Vanderplasschen
- Laboratory of Immunology-Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Avenue de Cureghem 10, B-4000 Liège, Belgium
| | - Benjamin G. Dewals
- Laboratory of Immunology-Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Avenue de Cureghem 10, B-4000 Liège, Belgium
- Laboratory of Parasitology, Faculty of Veterinary Medicine, FARAH, ULiège, Avenue de Cureghem 10, B-4000 Liège, Belgium
- Correspondence:
| |
Collapse
|
3
|
Bayliss RJ, Piguet V. Masters of manipulation: Viral modulation of the immunological synapse. Cell Microbiol 2018; 20:e12944. [PMID: 30123959 PMCID: PMC6492149 DOI: 10.1111/cmi.12944] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/01/2018] [Accepted: 08/14/2018] [Indexed: 02/06/2023]
Abstract
In order to thrive, viruses have evolved to manipulate host cell machinery for their own benefit. One major obstacle faced by pathogens is the immunological synapse. To enable efficient replication and latency in immune cells, viruses have developed a range of strategies to manipulate cellular processes involved in immunological synapse formation to evade immune detection and control T-cell activation. In vitro, viruses such as human immunodeficiency virus 1 and human T-lymphotropic virus type 1 utilise structures known as virological synapses to aid transmission of viral particles from cell to cell in a process termed trans-infection. The formation of the virological synapse provides a gateway for virus to be transferred between cells avoiding the extracellular space, preventing antibody neutralisation or recognition by complement. This review looks at how viruses are able to subvert intracellular signalling to modulate immune function to their advantage and explores the role synapse formation has in viral persistence and cell-to-cell transmission.
Collapse
Affiliation(s)
- Rebecca J. Bayliss
- Division of Infection and Immunity, School of MedicineCardiff UniversityCardiffUK
| | - Vincent Piguet
- Division of Infection and Immunity, School of MedicineCardiff UniversityCardiffUK
- Division of Dermatology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Division of DermatologyWomen's College HospitalTorontoOntarioCanada
| |
Collapse
|
4
|
Kwon EK, Min CK, Kim Y, Lee JW, Aigerim A, Schmidt S, Nam HJ, Han SK, Kim K, Cha JS, Kim H, Kim S, Cho HS, Choi MS, Cho NH. Constitutive activation of T cells by γ2-herpesviral GPCR through the interaction with cellular CXCR4. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:1-11. [PMID: 27751885 DOI: 10.1016/j.bbamcr.2016.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/14/2016] [Accepted: 10/11/2016] [Indexed: 12/28/2022]
Abstract
Members of the herpesviral family use multiple strategies to hijack infected host cells and exploit cellular signaling for their pathogenesis and latent infection. Among the most intriguing weapons in the arsenal of pathogenic herpesviruses are the constitutively active virally-encoded G protein-coupled receptors (vGPCRs). Even though vGPCRs contribute to viral pathogenesis such as immune evasion and proliferative disorders, the molecular details of how vGPCRs continuously activate cellular signaling are largely unknown. Here, we report that the vGPCR of Herpesvirus saimiri (HVS), an oncogenic γ2-herpesvirus, constitutively activates T cells via a heteromeric interaction with cellular CXCR4. Constitutive T cell activation also occurs with expression of the vGPCR of Kaposi's sarcoma-associated herpesvirus (KSHV), but not the vGPCR of Epstein-Barr virus. Expression of HVS vGPCR down-regulated the surface expression of CXCR4 but did not induce the degradation of the chemokine receptor, suggesting that vGPCR/CXCR4 signaling continues in cytosolic compartments. The physical association of vGPCR with CXCR4 was demonstrated by proximity ligation assay as well as immunoprecipitation. Interestingly, the constitutive activation of T cells by HVS vGPCR is independent of proximal T cell receptor (TCR) signaling molecules, such as TCRβ, Lck, and ZAP70, whereas CXCR4 silencing by shRNA abolished T cell activation by vGPCRs of HVS and KSHV. Furthermore, previously identified inactive vGPCR mutants failed to interact with CXCR4. These findings on the positive cooperativity of vGPCR with cellular CXCR4 in T cell activation extend our current understanding of the molecular mechanisms of vGPCR function and highlight the importance of heteromerization for GPCR activity.
Collapse
Affiliation(s)
- Eun-Kyung Kwon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Chan-Ki Min
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yuri Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jae-Won Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Abdimadiyeva Aigerim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sebastian Schmidt
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyun-Jun Nam
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Seong Kyu Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Kuglae Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jeong Seok Cha
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hoyoung Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Sanguk Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hyun-Soo Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Myung-Sik Choi
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Endemic Disease, Seoul National University Medical Research Center and Bundang Hospital, Seoul 03080, Republic of Korea.
| |
Collapse
|
5
|
Banerjee S, Uppal T, Strahan R, Dabral P, Verma SC. The Modulation of Apoptotic Pathways by Gammaherpesviruses. Front Microbiol 2016; 7:585. [PMID: 27199919 PMCID: PMC4847483 DOI: 10.3389/fmicb.2016.00585] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/11/2016] [Indexed: 12/11/2022] Open
Abstract
Apoptosis or programmed cell death is a tightly regulated process fundamental for cellular development and elimination of damaged or infected cells during the maintenance of cellular homeostasis. It is also an important cellular defense mechanism against viral invasion. In many instances, abnormal regulation of apoptosis has been associated with a number of diseases, including cancer development. Following infection of host cells, persistent and oncogenic viruses such as the members of the Gammaherpesvirus family employ a number of different mechanisms to avoid the host cell’s “burglar” alarm and to alter the extrinsic and intrinsic apoptotic pathways by either deregulating the expressions of cellular signaling genes or by encoding the viral homologs of cellular genes. In this review, we summarize the recent findings on how gammaherpesviruses inhibit cellular apoptosis via virus-encoded proteins by mediating modification of numerous signal transduction pathways. We also list the key viral anti-apoptotic proteins that could be exploited as effective targets for novel antiviral therapies in order to stimulate apoptosis in different types of cancer cells.
Collapse
Affiliation(s)
- Shuvomoy Banerjee
- Amity Institute of Virology and Immunology, Amity University Noida, India
| | - Timsy Uppal
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Roxanne Strahan
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Prerna Dabral
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Subhash C Verma
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| |
Collapse
|
6
|
Modulating p56Lck in T-Cells by a Chimeric Peptide Comprising Two Functionally Different Motifs of Tip from Herpesvirus saimiri. J Immunol Res 2015; 2015:395371. [PMID: 26539553 PMCID: PMC4619936 DOI: 10.1155/2015/395371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/28/2015] [Accepted: 05/27/2015] [Indexed: 11/18/2022] Open
Abstract
The Lck interacting protein Tip of Herpesvirus saimiri is responsible for T-cell transformation both in vitro and in vivo. Here we designed the chimeric peptide hTip-CSKH, comprising the Lck specific interacting motif CSKH of Tip and its hydrophobic transmembrane sequence (hTip), the latter as a vector targeting lipid rafts. We found that hTip-CSKH can induce a fivefold increase in proliferation of human and Aotus sp. T-cells. Costimulation with PMA did not enhance this proliferation rate, suggesting that hTip-CSKH is sufficient and independent of further PKC stimulation. We also found that human Lck phosphorylation was increased earlier after stimulation when T-cells were incubated previously with hTip-CSKH, supporting a strong signalling and proliferative effect of the chimeric peptide. Additionally, Lck downstream signalling was evident with hTip-CSKH but not with control peptides. Importantly, hTip-CSKH could be identified in heavy lipid rafts membrane fractions, a compartment where important T-cell signalling molecules (LAT, Ras, and Lck) are present during T-cell activation. Interestingly, hTip-CSKH was inhibitory to Jurkat cells, in total agreement with the different signalling pathways and activation requirements of this leukemic cell line. These results provide the basis for the development of new compounds capable of modulating therapeutic targets present in lipid rafts.
Collapse
|
7
|
Murid Gammaherpesvirus Latency-Associated Protein M2 Promotes the Formation of Conjugates between Transformed B Lymphoma Cells and T Helper Cells. PLoS One 2015; 10:e0142540. [PMID: 26544979 PMCID: PMC4636232 DOI: 10.1371/journal.pone.0142540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 10/22/2015] [Indexed: 12/02/2022] Open
Abstract
Establishment of persistent infection in memory B cells by murid herpesvirus-4 (MuHV-4) depends on the proliferation of latently infected germinal center B cells, for which T cell help is essential. Whether the virus is capable of modulating B-T helper cell interaction for its own benefit is still unknown. Here, we investigate if the MuHV-4 latency associated M2 protein, which assembles multiprotein complexes with B cell signaling proteins, plays a role. We observed that M2 led to the upregulation of adhesion and co-stimulatory molecules in transduced B cell lines. In an MHC-II restricted OVA peptide-specific system, M2 polarized to the B-T helper contact zone. Furthermore, it promoted B cell polarization, as demonstrated by the increased proximity of the B cell microtubule organizing center to the interface. Consistent with these data, M2 promoted the formation of B-T helper cell conjugates. In an in vitro competition assay, this translated into a competitive advantage, as T cells preferentially conjugated with M2-expressing B cells. However, expression of M2 alone in B cells was not sufficient to lead to T cell activation, as it only occurred in the presence of specific peptide. Taken together, these findings support that M2 promotes the formation of B-T helper cell conjugates. In an in vivo context this may confer a competitive advantage to the infected B cell in acquisition of T cell help and initiation of a germinal center reaction, hence host colonization.
Collapse
|
8
|
Species restriction of Herpesvirus saimiri and Herpesvirus ateles: Human lymphocyte transformation correlates with distinct signaling properties of viral oncoproteins. Virus Res 2012; 165:179-89. [DOI: 10.1016/j.virusres.2012.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 02/06/2012] [Accepted: 02/16/2012] [Indexed: 01/05/2023]
|
9
|
A molecular model for the differential activation of STAT3 and STAT6 by the herpesviral oncoprotein tip. PLoS One 2012; 7:e34306. [PMID: 22509288 PMCID: PMC3320567 DOI: 10.1371/journal.pone.0034306] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 02/27/2012] [Indexed: 01/27/2023] Open
Abstract
Constitutive STAT signaling provides growth promoting signals in many forms of malignancy. We performed molecular modeling and molecular dynamics studies of the interaction between the regulatory Src homology 2 (SH2) domains of STAT3 and 6 with phosphorylated peptides of the herpesviral oncoprotein Tip, which facilitates Src kinase mediated STAT-activation and T cell proliferation. The studies give insight into the ligand binding specificity of the STAT SH2 domains and provide the first model for the differential activation of STAT3 or STAT6 by two distinct regions of the viral Tip protein. The biological relevance of the modeled interactions was then confirmed by activation studies using corresponding recombinant oncoproteins, and finally by respective recombinant viruses. The functional data give experimental validation of the molecular dynamics study, and provide evidence for the involvement of STAT6 in the herpesvirus induced T cell proliferation.
Collapse
|
10
|
Katsch K, de Jong SJ, Albrecht JC, Steger J, Genth H, Posern G, Biesinger B. Actin-dependent activation of serum response factor in T cells by the viral oncoprotein tip. Cell Commun Signal 2012; 10:5. [PMID: 22385615 PMCID: PMC3310822 DOI: 10.1186/1478-811x-10-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 03/03/2012] [Indexed: 01/05/2023] Open
Abstract
Serum response factor (SRF) acts as a multifunctional transcription factor regulated by mutually exclusive interactions with ternary complex factors (TCFs) or myocardin-related transcription factors (MRTFs). Binding of Rho- and actin-regulated MRTF:SRF complexes to target gene promoters requires an SRF-binding site only, whereas MAPK-regulated TCF:SRF complexes in addition rely on flanking sequences present in the serum response element (SRE). Here, we report on the activation of an SRE luciferase reporter by Tip, the viral oncoprotein essentially contributing to human T-cell transformation by Herpesvirus saimiri. SRE activation in Tip-expressing Jurkat T cells could not be attributed to triggering of the MAPK pathway. Therefore, we further analyzed the contribution of MRTF complexes. Indeed, Tip also activated a reporter construct responsive to MRTF:SRF. Activation of this reporter was abrogated by overexpression of a dominant negative mutant of the MRTF-family member MAL. Moreover, enrichment of monomeric actin suppressed the Tip-induced reporter activity. Further upstream, the Rho-family GTPase Rac, was found to be required for MRTF:SRF reporter activation by Tip. Initiation of this pathway was strictly dependent on Tip's ability to interact with Lck and on the activity of this Src-family kinase. Independent of Tip, T-cell stimulation orchestrates Src-family kinase, MAPK and actin pathways to induce SRF. These findings establish actin-regulated transcription in human T cells and suggest its role in viral oncogenesis.
Collapse
Affiliation(s)
- Kristin Katsch
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | | | | | | |
Collapse
|
11
|
Kliem C, Merling A, Giaisi M, Köhler R, Krammer PH, Li-Weber M. Curcumin suppresses T cell activation by blocking Ca2+ mobilization and nuclear factor of activated T cells (NFAT) activation. J Biol Chem 2012; 287:10200-10209. [PMID: 22303019 DOI: 10.1074/jbc.m111.318733] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Curcumin is the active ingredient of the spice turmeric and has been shown to have a number of pharmacologic and therapeutic activities including antioxidant, anti-microbial, anti-inflammatory, and anti-carcinogenic properties. The anti-inflammatory effects of curcumin have primarily been attributed to its inhibitory effect on NF-κB activity due to redox regulation. In this study, we show that curcumin is an immunosuppressive phytochemical that blocks T cell-activation-induced Ca(2+) mobilization with IC(50) = ∼12.5 μM and thereby prevents NFAT activation and NFAT-regulated cytokine expression. This finding provides a new mechanism for curcumin-mediated anti-inflammatory and immunosuppressive function. We also show that curcumin can synergize with CsA to enhance immunosuppressive activity because of different inhibitory mechanisms. Furthermore, because Ca(2+) is also the secondary messenger crucial for the TCR-induced NF-κB signaling pathway, our finding also provides another mechanism by which curcumin suppresses NF-κB activation.
Collapse
Affiliation(s)
- Christian Kliem
- Technologietransfer (T010), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Anette Merling
- Tumorimmunology Program (D030), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Marco Giaisi
- Tumorimmunology Program (D030), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Rebecca Köhler
- Tumorimmunology Program (D030), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Peter H Krammer
- Tumorimmunology Program (D030), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Min Li-Weber
- Tumorimmunology Program (D030), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany.
| |
Collapse
|
12
|
Inhibition of human papillomavirus DNA replication by an E1-derived p80/UAF1-binding peptide. J Virol 2012; 86:3486-500. [PMID: 22278251 DOI: 10.1128/jvi.07003-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The papillomavirus E1 helicase is recruited by E2 to the viral origin, where it assembles into a double hexamer that orchestrates replication of the viral genome. We previously identified the cellular WD40 repeat-containing protein p80/UAF1 as a novel interaction partner of E1 from anogenital human papillomavirus (HPV) types. p80 was found to interact with the first 40 residues of HPV type 31 (HPV31) E1, and amino acid substitutions within this domain abrogated the maintenance of the viral episome in keratinocytes. In this study, we report that these p80-binding substitutions reduce by 70% the ability of E1 to support transient viral DNA replication without affecting its interaction with E2 and assembly at the origin in vivo. Microscopy studies revealed that p80 is relocalized from the cytoplasm to discrete subnuclear foci by E1 and E2. Chromatin immunoprecipitation assays further revealed that p80 is recruited to the viral origin in an E1- and E2-dependent manner. Interestingly, overexpression of a 40-amino-acid-long p80-binding peptide, derived from HPV31 E1, was found to inhibit viral DNA replication by preventing the recruitment of endogenous p80 to the origin. Mutant peptides defective for p80 interaction were not inhibitory, demonstrating the specificity of this effect. Characterization of this E1 peptide by nuclear magnetic resonance (NMR) showed that it is intrinsically disordered in solution, while mapping studies indicated that the WD repeats of p80 are required for E1 interaction. These results provide additional evidence for the requirement for p80 in anogenital HPV DNA replication and highlight the potential of E1-p80 interaction as a novel antiviral target.
Collapse
|
13
|
Loss-of-function mutations within the IL-2 inducible kinase ITK in patients with EBV-associated lymphoproliferative diseases. Leukemia 2012; 26:963-71. [PMID: 22289921 DOI: 10.1038/leu.2011.371] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The purpose of this study was the appraisal of the clinical and functional consequences of germline mutations within the gene for the IL-2 inducible T-cell kinase, ITK. Among patients with Epstein-Barr virus-driven lymphoproliferative disorders (EBV-LPD), negative for mutations in SH2D1A and XIAP (n=46), we identified two patients with R29H or D500T,F501L,M503X mutations, respectively. Human wild-type (wt) ITK, but none of the mutants, was able to rescue defective calcium flux in murine Itk(-/-) T cells. Pulse-chase experiments showed that ITK mutations lead to varying reductions of protein half-life from 25 to 69% as compared with wt ITK (107 min). The pleckstrin homology domain of wt ITK binds most prominently to phosphatidylinositol monophosphates (PI(3)P, PI(4)P, PI(5)P) and to lesser extend to its double or triple phosphorylated derivates (PIP2, PIP3), interactions which were dramatically reduced in the patient with the ITK(R29H) mutant. ITK mutations are distributed over the entire protein and include missense, nonsense and indel mutations, reminiscent of the situation in its sister kinase in B cells, Bruton's tyrosine kinase.
Collapse
|
14
|
Kim Y, Kwon EK, Jeon JH, So I, Kim IG, Choi MS, Kim IS, Choi JK, Jung JU, Cho NH. Activation of the STAT6 transcription factor in Jurkat T-cells by the herpesvirus saimiri Tip protein. J Gen Virol 2011; 93:330-340. [PMID: 22012462 DOI: 10.1099/vir.0.036087-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Herpesvirus saimiri (HVS), a T-lymphotropic monkey herpesvirus, induces fulminant T-cell lymphoma in non-natural primate hosts. In addition, it can immortalize human T-cells in vitro. HVS tyrosine kinase-interacting protein (Tip) is an essential viral gene required for T-cell transformation both in vitro and in vivo. In this study, we found that Tip interacts with the STAT6 transcription factor and induces phosphorylation of STAT6 in T-cells. The interaction with STAT6 requires the Tyr(127) residue and Lck-binding domain of Tip, which are indispensable for interleukin (IL)-2-independent T-cell transformation by HVS. It was also demonstrated that Tip induces nuclear translocation of STAT6, as well as activation of STAT6-dependent transcription in Jurkat T-cells. Interestingly, the phosphorylated STAT6 mainly colocalized with vesicles containing Tip within T-cells, but was barely detectable in the nucleus. However, nuclear translocation of phospho-STAT6 and transcriptional activation of STAT6 by IL-4 stimulation were not affected significantly in T-cells expressing Tip. Collectively, these findings suggest that the constitutive activation of STAT6 by Tip in T-cells may contribute to IL-2-independent T-cell transformation by HVS.
Collapse
Affiliation(s)
- Yuri Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun-Kyung Kwon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - In-Gyu Kim
- Department of Biochemistry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myung-Sik Choi
- Institute of Endemic Disease, Seoul National University Medical Research Center and Bundang Hospital, Jongno-Gu, Seoul 110-799, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ik-Sang Kim
- Institute of Endemic Disease, Seoul National University Medical Research Center and Bundang Hospital, Jongno-Gu, Seoul 110-799, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joong-Kook Choi
- Division of Biochemistry, College of Medicine, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | - Jae Ung Jung
- Molecular Microbiology and Immunology, University of Southern California, School of Medicine, 2011 Zonal Avenue, HMR401, Los Angeles, CA 90033, USA
| | - Nam-Hyuk Cho
- Institute of Endemic Disease, Seoul National University Medical Research Center and Bundang Hospital, Jongno-Gu, Seoul 110-799, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Kingston D, Chang H, Ensser A, Lee HR, Lee J, Lee SH, Jung JU, Cho NH. Inhibition of retromer activity by herpesvirus saimiri tip leads to CD4 downregulation and efficient T cell transformation. J Virol 2011; 85:10627-38. [PMID: 21849449 PMCID: PMC3187508 DOI: 10.1128/jvi.00757-11] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 08/08/2011] [Indexed: 11/20/2022] Open
Abstract
The mammalian retromer is an evolutionally conserved protein complex composed of a vacuolar protein sorting trimer (Vps 26/29/35) that participates in cargo recognition and a sorting nexin (SNX) dimer that binds to endosomal membranes. The retromer plays an important role in efficient retrograde transport for endosome-to-Golgi retrieval of the cation-independent mannose-6-phosphate receptor (CI-MPR), a receptor for lysosomal hydrolases, and other endosomal proteins. This ultimately contributes to the control of cell growth, cell adhesion, and cell migration. The herpesvirus saimiri (HVS) tyrosine kinase-interacting protein (Tip), required for the immortalization of primary T lymphocytes, targets cellular signaling molecules, including Lck tyrosine kinases and the p80 endosomal trafficking protein. Despite the pronounced effects of HVS Tip on T cell signal transduction, the details of its activity on T cell immortalization remain elusive. Here, we report that the amino-terminal conserved, glutamate-rich sequence of Tip specifically interacts with the retromer subunit Vps35 and that this interaction not only causes the redistribution of Vps35 from the early endosome to the lysosome but also drastically inhibits retromer activity, as measured by decreased levels of CI-MPR and lower activities of cellular lysosomal hydrolases. Physiologically, the inhibition of intracellular retromer activity by Tip is ultimately linked to the downregulation of CD4 surface expression and to the efficient in vitro immortalization of primary human T cells to interleukin-2 (IL-2)-independent permanent growth. Therefore, HVS Tip uniquely targets the retromer complex to impair the intracellular trafficking functions of infected cells, ultimately contributing to efficient T cell transformation.
Collapse
Affiliation(s)
- Dior Kingston
- Department of Microbiology and Molecular Genetics and Tumor Virology Division, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
- Institute for Research in Biomedicine, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Heesoon Chang
- Department of Microbiology and Molecular Genetics and Tumor Virology Division, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
- Molecular Microbiology & Immunology, University of Southern California, School of Medicine, 2011 Zonal Avenue, HMR401, Los Angeles, California 90033
| | - Armin Ensser
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Hye-Ra Lee
- Department of Microbiology and Molecular Genetics and Tumor Virology Division, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
- Molecular Microbiology & Immunology, University of Southern California, School of Medicine, 2011 Zonal Avenue, HMR401, Los Angeles, California 90033
| | - Jongsoo Lee
- Department of Microbiology and Molecular Genetics and Tumor Virology Division, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
- Molecular Microbiology & Immunology, University of Southern California, School of Medicine, 2011 Zonal Avenue, HMR401, Los Angeles, California 90033
| | - Sun-Hwa Lee
- Molecular Microbiology & Immunology, University of Southern California, School of Medicine, 2011 Zonal Avenue, HMR401, Los Angeles, California 90033
| | - Jae Ung Jung
- Department of Microbiology and Molecular Genetics and Tumor Virology Division, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
- Molecular Microbiology & Immunology, University of Southern California, School of Medicine, 2011 Zonal Avenue, HMR401, Los Angeles, California 90033
| | - Nam-Hyuk Cho
- Department of Microbiology and Molecular Genetics and Tumor Virology Division, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
- Department of Microbiology and Immunology, Seoul National University College of Medicine, and Institute of Endemic Disease, Seoul National University Medical Research Center and Bundang Hospital, Jongno-Gu, Seoul 110-799, Republic of Korea
| |
Collapse
|
16
|
Van Prooyen N, Gold H, Andresen V, Schwartz O, Jones K, Ruscetti F, Lockett S, Gudla P, Venzon D, Franchini G. Human T-cell leukemia virus type 1 p8 protein increases cellular conduits and virus transmission. Proc Natl Acad Sci U S A 2010; 107:20738-43. [PMID: 21076035 PMCID: PMC2996430 DOI: 10.1073/pnas.1009635107] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The human T-cell leukemia virus type 1 (HTLV-1) is the cause of adult T-cell leukemia/lymphoma as well as tropical spastic paraparesis/HTLV-1-associated myelopathy. HTLV-1 is transmitted to T cells through the virological synapse and by extracellular viral assemblies. Here, we uncovered an additional mechanism of virus transmission that is regulated by the HTLV-1-encoded p8 protein. We found that the p8 protein, known to anergize T cells, is also able to increase T-cell contact through lymphocyte function-associated antigen-1 clustering. In addition, p8 augments the number and length of cellular conduits among T cells and is transferred to neighboring T cells through these conduits. p8, by establishing a T-cell network, enhances the envelope-dependent transmission of HTLV-1. Thus, the ability of p8 to simultaneously anergize and cluster T cells, together with its induction of cellular conduits, secures virus propagation while avoiding the host's immune surveillance. This work identifies p8 as a viral target for the development of therapeutic strategies that may limit the expansion of infected cells in HTLV-1 carriers and decrease HTLV-1-associated morbidity.
Collapse
Affiliation(s)
- Nancy Van Prooyen
- Animal Models Retroviral Vaccine Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
- Department of Biology, John Hopkins University, Baltimore, MD 21218
| | - Heather Gold
- Animal Models Retroviral Vaccine Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Vibeke Andresen
- Animal Models Retroviral Vaccine Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Owen Schwartz
- Biological Imaging, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kathryn Jones
- Basic Research Program, Science Applications International Corporation–Frederick, National Cancer Institute–Frederick, Frederick, MD 21702
| | - Frank Ruscetti
- Laboratory of Experimental Immunology, National Cancer Institute–Frederick, Frederick, MD 21702
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, National Cancer Institute–Frederick, Frederick MD 21702
| | - Prabhakar Gudla
- Image Analysis Laboratory, Research Tech Program, Science Applications International Corporation–Frederick, National Cancer Institute–Frederick, Frederick, MD 21702; and
| | - David Venzon
- Biostatistics and Data Management Section, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Genoveffa Franchini
- Animal Models Retroviral Vaccine Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
17
|
Vesicle traffic to the immunological synapse: a multifunctional process targeted by lymphotropic viruses. Curr Top Microbiol Immunol 2010; 340:191-207. [PMID: 19960315 DOI: 10.1007/978-3-642-03858-7_10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The site of contact between T lymphocytes and antigen-presenting cells becomes, upon antigen recognition, an organized junction named the immunological synapse. Various T cell organelles polarize, together with microtubules, toward the antigen-presenting cell. Among them, intracellular vesicular compartments, such as the Golgi apparatus, the recycling endosomal compartment, or cytotoxic granules help to build the immunological synapse and ensure effector functions, such as polarized secretion of cytokines by helper T cells, or exocytosis of lytic granules by cytotoxic T cells. Lymphotropic retroviruses, such as the human immunodeficiency virus type 1, the human T cell leukemia virus type 1, or the Herpesvirus saimiri, can subvert some of the vesicle traffic mechanisms impeding the generation and function of the immunological synapses. This review focuses on the polarization of vesicle traffic, its regulation, and its role in maintaining the structure and function of the immunological synapse. We discuss how some lymphotropic viruses target the vesicle traffic in T lymphocytes, inhibiting the formation of immunological synapses and modulating the response of infected T cells.
Collapse
|
18
|
Arhel N, Lehmann M, Clauss K, Nienhaus GU, Piguet V, Kirchhoff F. The inability to disrupt the immunological synapse between infected human T cells and APCs distinguishes HIV-1 from most other primate lentiviruses. J Clin Invest 2009; 119:2965-75. [PMID: 19759518 DOI: 10.1172/jci38994] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 07/22/2009] [Indexed: 12/31/2022] Open
Abstract
Viruses that infect T cells, including those of the lentivirus genus, such as HIV-1, modulate the responsiveness of infected T cells to stimulation by interacting APCs in a manner that renders the T cells more permissive for viral replication. HIV-1 and other primate lentiviruses use their Nef proteins to manipulate the T cell/APC contact zone, the immunological synapse (IS). It is known that primate lentiviral Nef proteins differ substantially in their ability to modulate cell surface expression of the TCR-CD3 and CD28 receptors critical for the formation and function of the IS. However, the impact of these differences in Nef function on the interaction and communication between virally infected T cells and primary APCs has not been investigated. Here we have used primary human cells to show that Nef proteins encoded by HIV-2 and most SIVs, which downmodulate cell surface expression of TCR-CD3, disrupt formation of the IS between infected T cells and Ag-presenting macrophages or DCs. In contrast, nef alleles from HIV-1 and its simian precursor SIVcpz failed to suppress synapse formation and events downstream of TCR signaling. Our data suggest that most primate lentiviruses disrupt communication between virally infected CD4+ Th cells and APCs, whereas HIV-1 and its SIV precursor have largely lost this capability. The resulting differences in the levels of T cell activation and apoptosis may play a role in the pathogenesis of AIDS.
Collapse
Affiliation(s)
- Nathalie Arhel
- Institute of Molecular Virology, University of Ulm, Albert-Einstein-Allee 11, Ulm, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Mycobacterium tuberculosis cell wall glycolipids directly inhibit CD4+ T-cell activation by interfering with proximal T-cell-receptor signaling. Infect Immun 2009; 77:4574-83. [PMID: 19651854 DOI: 10.1128/iai.00222-09] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Immune evasion is required for Mycobacterium tuberculosis to survive in the face of robust adaptive CD4(+) T-cell responses. We have previously shown that M. tuberculosis can indirectly inhibit CD4(+) T cells by suppressing the major histocompatibility complex class II antigen-presenting cell function of macrophages. This study was undertaken to determine if M. tuberculosis could directly inhibit CD4(+) T-cell activation. Murine CD4(+) T cells were purified from spleens by negative immunoaffinity selection followed by flow sorting. Purified CD4(+) T cells were activated for 16 to 48 h with CD3 and CD28 monoclonal antibodies in the presence or absence of M. tuberculosis and its subcellular fractions. CD4(+) T-cell activation was measured by interleukin 2 production, proliferation, and expression of activation markers, all of which were decreased in the presence of M. tuberculosis. Fractionation identified that M. tuberculosis cell wall glycolipids, specifically, phosphatidylinositol mannoside and mannose-capped lipoarabinomannan, were potent inhibitors. Glycolipid-mediated inhibition was not dependent on Toll-like receptor signaling and could be bypassed through stimulation with phorbol 12-myristate 13-acetate and ionomycin. ZAP-70 phosphorylation was decreased in the presence of M. tuberculosis glycolipids, indicating that M. tuberculosis glycolipids directly inhibited CD4(+) T-cell activation by interfering with proximal T-cell-receptor signaling.
Collapse
|
20
|
Haller C, Fackler OT. HIV-1 at the immunological and T-lymphocytic virological synapse. Biol Chem 2009; 389:1253-60. [PMID: 18713012 DOI: 10.1515/bc.2008.143] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cell-cell transmission of human immunodeficiency virus type 1 (HIV-1) is considered the most effective mode of viral spread in T-lymphocyte cultures. Evidence has accumulated that HIV-1 assembles polarized synaptic-like structures, referred to as virological synapses, as specialized sites of viral transfer. Interestingly, it was recently also discovered that HIV-1 impairs the formation of the structurally similar immunological synapse, thereby modulating exogenous T-lymphocyte stimulation to yield an optimal activation state for productive HIV-1 infection. The careful dissection of these opposing effects will contribute to our understanding of retroviral spread and cellular signal transduction machineries.
Collapse
Affiliation(s)
- Claudia Haller
- Department of Virology, University of Heidelberg, INF 324, D-69120 Heidelberg, Germany
| | | |
Collapse
|
21
|
Min CK, Bang SY, Cho BA, Choi YH, Yang JS, Lee SH, Seong SY, Kim KW, Kim S, Jung JU, Choi MS, Kim IS, Cho NH. Role of amphipathic helix of a herpesviral protein in membrane deformation and T cell receptor downregulation. PLoS Pathog 2008; 4:e1000209. [PMID: 19023411 PMCID: PMC2581436 DOI: 10.1371/journal.ppat.1000209] [Citation(s) in RCA: 23] [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: 05/30/2008] [Accepted: 10/16/2008] [Indexed: 11/18/2022] Open
Abstract
Lipid rafts are membrane microdomains that function as platforms for signal transduction and membrane trafficking. Tyrosine kinase interacting protein (Tip) of T lymphotropic Herpesvirus saimiri (HVS) is targeted to lipid rafts in T cells and downregulates TCR and CD4 surface expression. Here, we report that the membrane-proximal amphipathic helix preceding Tip's transmembrane (TM) domain mediates lipid raft localization and membrane deformation. In turn, this motif directs Tip's lysosomal trafficking and selective TCR downregulation. The amphipathic helix binds to the negatively charged lipids and induces liposome tubulation, the TM domain mediates oligomerization, and cooperation of the membrane-proximal helix with the TM domain is sufficient for localization to lipid rafts and lysosomal compartments, especially the mutivesicular bodies. These findings suggest that the membrane-proximal amphipathic helix and TM domain provide HVS Tip with the unique ability to deform the cellular membranes in lipid rafts and to downregulate TCRs potentially through MVB formation.
Collapse
Affiliation(s)
- Chan-Ki Min
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
| | - Sun-Young Bang
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
| | - Bon-A Cho
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
| | - Yun-Hui Choi
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
| | - Jae-Seong Yang
- Department of Life Science and School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Kyungbuk, Korea
| | - Sun-Hwa Lee
- Seoul National University Hospital, Innovative Research Institute for Cell Therapy, Chongno-Gu, Seoul, Korea
| | - Seung-Yong Seong
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
- Seoul National University Hospital, Innovative Research Institute for Cell Therapy, Chongno-Gu, Seoul, Korea
| | - Ki Woo Kim
- National Instrumentation Center for Environmental Management, Seoul National University, Gwanak-Gu, Seoul, Korea
| | - Sanguk Kim
- Department of Life Science and School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Kyungbuk, Korea
| | - Jae Ung Jung
- Department of Molecular Microbiology and Immunology, University of Southern California School of Medicine, Los Angeles, California, United States of America
| | - Myung-Sik Choi
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
| | - Ik-Sang Kim
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, College of Medicine and Institute of Endemic Diseases, Seoul National University Medical Research Center and Bundang Hospital, Seoul, Korea
- * E-mail:
| |
Collapse
|
22
|
González PA, Prado CE, Leiva ED, Carreño LJ, Bueno SM, Riedel CA, Kalergis AM. Respiratory syncytial virus impairs T cell activation by preventing synapse assembly with dendritic cells. Proc Natl Acad Sci U S A 2008; 105:14999-5004. [PMID: 18818306 PMCID: PMC2567482 DOI: 10.1073/pnas.0802555105] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Indexed: 12/11/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection is one of the leading causes of infant hospitalization and a major health and economic burden worldwide. Infection with this virus induces an exacerbated innate proinflammatory immune response characterized by abundant immune cell infiltration into the airways and lung tissue damage. RSV also impairs the induction of an adequate adaptive T cell immune response, which favors virus pathogenesis. Unfortunately, to date there are no efficient vaccines against this virus. Recent in vitro and in vivo studies suggest that RSV infection can prevent T cell activation, a phenomenon attributed in part to cytokines and chemokines secreted by RSV-infected cells. Efficient immunity against viruses is promoted by dendritic cells (DCs), professional antigen-presenting cells, that prime antigen-specific helper and cytotoxic T cells. Therefore, it would be to the advantage of RSV to impair DC function and prevent the induction of T cell immunity. Here, we show that, although RSV infection induces maturation of murine DCs, these cells are rendered unable to activate antigen-specific T cells. Inhibition of T cell activation by RSV was observed independently of the type of TCR ligand on the DC surface and applied to cognate-, allo-, and superantigen stimulation. As a result of exposure to RSV-infected DCs, T cells became unresponsive to subsequent TCR engagement. RSV-mediated impairment in T cell activation required DC-T cell contact and involved inhibition of immunological synapse assembly among these cells. Our data suggest that impairment of immunological synapse could contribute to RSV pathogenesis by evading adaptive immunity and reducing T cell-mediated virus clearance.
Collapse
Affiliation(s)
- Pablo A. González
- *Millennium Nucleus of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, and
| | - Carolina E. Prado
- *Millennium Nucleus of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, and
| | - Eduardo D. Leiva
- *Millennium Nucleus of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, and
| | - Leandro J. Carreño
- *Millennium Nucleus of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, and
| | - Susan M. Bueno
- *Millennium Nucleus of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, and
| | - Claudia A. Riedel
- Laboratorio de Biología Celular y Farmacología, Departamento de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Alexis M. Kalergis
- *Millennium Nucleus of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, and
- Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile; and
| |
Collapse
|
23
|
NF-kappaB activation by the viral oncoprotein StpC enhances IFN-gamma production in T cells. Immunol Cell Biol 2008; 86:622-30. [PMID: 18560378 DOI: 10.1038/icb.2008.43] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interferon-gamma (IFN-gamma) is an essential regulator of innate and adaptive immune responses and a hallmark of the Th1 T-cell subset. It is produced at high levels by human T lymphocytes upon transformation with Herpesvirus saimiri, which depends on the expression of the viral oncoproteins saimiri transformation-associated protein of subgroup C (StpC) and tyrosine kinase-interacting protein (Tip). Here, we show that IFN-gamma production was induced by Tip in Jurkat T cells. StpC by itself did not affect IFN-gamma expression, but enhanced the effect of Tip. Our results substantiated the findings that StpC induces NF-kappaB activation and demonstrated that other transcription factors, including NFAT, AP-1 and serum response element regulators, were not activated by StpC in unstimulated T cells. Studies using StpC mutants deficient in NF-kappaB activation, dominant negative IkappaBalpha and constitutively active IKK2, established the importance of NF-kappaB in StpC-mediated upregulation of IFN-gamma production. These observations suggest that NF-kappaB induction by StpC contributes to the Th1-like phenotype of virus-transformed human T cells.
Collapse
|
24
|
|
25
|
Fukumoto R, Dundr M, Nicot C, Adams A, Valeri VW, Samelson LE, Franchini G. Inhibition of T-cell receptor signal transduction and viral expression by the linker for activation of T cells-interacting p12(I) protein of human T-cell leukemia/lymphoma virus type 1. J Virol 2007; 81:9088-99. [PMID: 17582004 PMCID: PMC1951423 DOI: 10.1128/jvi.02703-06] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The p12(I) protein of human T-cell leukemia/lymphoma virus type 1 (HTLV-1) is a small oncoprotein that increases calcium release following protein kinase C activation by phorbol myristate acetate, and importantly, this effect is linker for activation of T cells (LAT) independent. Here, we demonstrate that p12(I) inhibits the phosphorylation of LAT, Vav, and phospholipase C-gamma 1 and decreases NFAT (nuclear factor of activated T cells) activation upon engagement of the T-cell receptor (TCR) with anti-CD3 antibody. Furthermore, we demonstrate that p12(I) localizes to membrane lipid rafts and, upon engagement of the TCR, relocalizes to the interface between T cells and antigen-presenting cells, defined as the immunological synapse. A p12(I) knockout molecular clone of HTLV-1 expresses more virus upon antigen stimulation than the isogenic wild type, suggesting that, by decreasing T-cell responsiveness, p12(I) curtails viral expression. Thus, p12(I) has contrasting effects on TCR signaling: it down-regulates TCR in a LAT-dependent manner on one hand, and on the other, it increases calcium release in a LAT-independent manner. The negative regulation of T-cell activation by p12(I) may have evolved to minimize immune recognition of infected CD4(+) T cells, to impair the function of infected cytotoxic CD8(+) T cells, and to favor viral persistence in the infected host.
Collapse
Affiliation(s)
- Risaku Fukumoto
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, NIH, Bethesda, MD 20892-5065, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Cho NH, Kingston D, Chang H, Kwon EK, Kim JM, Lee JH, Chu H, Choi MS, Kim IS, Jung JU. Association of herpesvirus saimiri tip with lipid raft is essential for downregulation of T-cell receptor and CD4 coreceptor. J Virol 2007; 80:108-18. [PMID: 16352535 PMCID: PMC1317525 DOI: 10.1128/jvi.80.1.108-118.2006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lipid rafts are membrane microdomains that are proposed to function as platforms for both receptor signaling and trafficking. Our previous studies have demonstrated that Tip of herpesvirus saimiri (HVS), which is a T-lymphotropic tumor virus, is constitutively targeted to lipid rafts and interacts with cellular Lck tyrosine kinase and p80 WD repeat-containing endosomal protein. Through the interactions with Lck and p80, HVS Tip modulates diverse T-cell functions, which leads to the downregulation of T-cell receptor (TCR) and CD4 coreceptor surface expression, the inhibition of TCR signal transduction, and the activation of STAT3 transcription factor. In this study, we investigated the functional significance of Tip association with lipid rafts. We found that Tip expression remarkably increased lipid raft fractions in human T cells by enhancing the recruitment of lipid raft-resident proteins. Genetic analysis showed that the carboxyl-terminal transmembrane, but not p80 and Lck interaction, of Tip was required for the lipid raft localization and that lipid raft localization of Tip was necessary for the efficient downregulation of TCR and CD4 surface expression. Correlated with this, treatment with Filipin III, a lipid raft-disrupting agent, effectively reversed the downregulation of CD3 and CD4 surface expression induced by Tip. On the other hand, Tip mutants that were no longer present in lipid rafts were still capable of inhibiting TCR signaling and activating STAT3 transcription factor activity as efficiently as wild-type (wt) Tip. These results indicate that the association of Tip with lipid rafts is essential for the downregulation of TCR and CD4 surface expression but not for the inhibition of TCR signal transduction and the activation of STAT3 transcription factor. These results also suggest that the signaling and targeting activities of HVS Tip rely on functionally and genetically separable mechanisms, which may independently modulate T-cell function for viral persistence or pathogenesis.
Collapse
Affiliation(s)
- Nam-Hyuk Cho
- Department of Microbiology and Molecular Genetics and Tumor Virology Division, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Heck E, Friedrich U, Gack MU, Lengenfelder D, Schmidt M, Müller-Fleckenstein I, Fleckenstein B, Ensser A, Biesinger B. Growth transformation of human T cells by herpesvirus saimiri requires multiple Tip-Lck interaction motifs. J Virol 2006; 80:9934-42. [PMID: 17005671 PMCID: PMC1617286 DOI: 10.1128/jvi.01112-06] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lymphoma induction and T-cell transformation by herpesvirus saimiri strain C488 depends on two viral oncoproteins, StpC and Tip. The major interaction partner of Tip is the protein tyrosine kinase Lck, a key regulator of T-cell activation. The Lck binding domain (LBD) of Tip comprises two interaction motifs, a proline-rich SH3 domain-binding sequence (SH3B) and a region with homology to the C terminus of Src family kinase domains (CSKH). In addition, biophysical binding analyses with purified Lck-SH2 domain suggest the phosphorylated tyrosine residue 127 of Tip (pY127) as a potential third Lck interaction site. Here, we addressed the relevance of the individual binding motifs, SH3B, CSKH, and pY127, for Tip-Lck interaction and for human T-cell transformation. Both motifs within the LBD displayed Lck binding activities and cooperated to achieve a highly efficient interaction, while pY127, the major tyrosine phosphorylation site of Tip, did not enhance Lck binding in T cells. Herpesvirus saimiri strain C488 recombinants lacking one or both LBD motifs of Tip lost their transforming potential on human cord blood lymphocytes. Recombinant virus expressing Tip with a mutation at position Y127 was still able to transform human T lymphocytes but, in contrast to wild-type virus, was strictly dependent on exogenous interleukin-2. Thus, the strong Lck binding mediated by cooperation of both LBD motifs was essential for the transformation of human T cells by herpesvirus saimiri C488. The major tyrosine phosphorylation site Y127 of Tip was particularly required for transformation in the absence of exogenous interleukin-2, suggesting its involvement in cytokine signaling pathways.
Collapse
Affiliation(s)
- Elke Heck
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schlossgarten 4, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Brinkmann MM, Schulz TF. Regulation of intracellular signalling by the terminal membrane proteins of members of the Gammaherpesvirinae. J Gen Virol 2006; 87:1047-1074. [PMID: 16603506 DOI: 10.1099/vir.0.81598-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The human gamma(1)-herpesvirus Epstein-Barr virus (EBV) and the gamma(2)-herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV), rhesus rhadinovirus (RRV), herpesvirus saimiri (HVS) and herpesvirus ateles (HVA) all contain genes located adjacent to the terminal-repeat region of their genomes, encoding membrane proteins involved in signal transduction. Designated 'terminal membrane proteins' (TMPs) because of their localization in the viral genome, they interact with a variety of cellular signalling molecules, such as non-receptor protein tyrosine kinases, tumour-necrosis factor receptor-associated factors, Ras and Janus kinase (JAK), thereby initiating further downstream signalling cascades, such as the MAPK, PI3K/Akt, NF-kappaB and JAK/STAT pathways. In the case of TMPs expressed during latent persistence of EBV and HVS (LMP1, LMP2A, Stp and Tip), their modulation of intracellular signalling pathways has been linked to the provision of survival signals to latently infected cells and, hence, a contribution to occasional cellular transformation. In contrast, activation of similar pathways by TMPs of KSHV (K1 and K15) and RRV (R1), expressed during lytic replication, may extend the lifespan of virus-producing cells, alter their migration and/or modulate antiviral immune responses. Whether R1 and K1 contribute to the oncogenic properties of KSHV and RRV has not been established satisfactorily, despite their transforming qualities in experimental settings.
Collapse
Affiliation(s)
- Melanie M Brinkmann
- Institut für Virologie, Medizinische Hochschule Hannover, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| | - Thomas F Schulz
- Institut für Virologie, Medizinische Hochschule Hannover, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
29
|
Sloan DD, Han JY, Sandifer TK, Stewart M, Hinz AJ, Yoon M, Johnson DC, Spear PG, Jerome KR. Inhibition of TCR signaling by herpes simplex virus. THE JOURNAL OF IMMUNOLOGY 2006; 176:1825-33. [PMID: 16424213 DOI: 10.4049/jimmunol.176.3.1825] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T lymphocytes are an essential component of the immune response against HSV infection. We previously reported that T cells became functionally impaired or inactivated after contacting HSV-infected fibroblasts. In our current study, we investigate the mechanisms of inactivation. We report that HSV-infected fibroblasts or HSV alone can inactivate T cells by profoundly inhibiting TCR signal transduction. Inactivation requires HSV penetration into T cells but not de novo transcription or translation. In HSV-inactivated T cells stimulated through the TCR, phosphorylation of Zap70 occurs normally. However, TCR signaling is inhibited at linker for activation of T cells (LAT) and at steps distal to LAT in the TCR signal cascade including inhibition of calcium flux and inhibition of multiple MAPK. Inactivation of T cells by HSV leads to the reduced phosphorylation of LAT at tyrosine residues critical for TCR signal propagation. Treatment of T cells with tyrosine phosphatase inhibitors attenuates inactivation by HSV, and stimulus with a mitogen that bypasses LAT phosphorylation overcomes inactivation. Our findings elucidate a potentially novel method of viral immune evasion that could be exploited to better manage HSV infection, aid in vaccine design, or allow targeted manipulation of T cell function.
Collapse
Affiliation(s)
- Derek D Sloan
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Acute T-lymphoproliferative syndromes are caused by herpesvirus saimiri (HVS) and ateles in neotropical primates; by alcelaphine herpesvirus-1 and ovine herpesvirus-2 strains in domestic cattle and other ungulates; and by the α-herpesvirus of Marek's disease in chickens. T-cell lymphoproliferation caused by these herpesviruses has short incubation periods and a rapid course when compared with retroviral disease. The B-lymphotropic Epstein–Barr virus (EBV) is also associated with some human T-cell malignancies. Analogous to EBV in B cells, HVS isolates of the subgroup C are uniquely capable of transforming human and Old World primate T lymphocytes to continuous growth in cell culture and can provide useful tools for T-cell immunology or gene transfer. Signal transduction pathways stimulated by the viral oncoproteins seem to converge at related cellular effector proteins, in total providing a proproliferative signal. However, the viral oncoproteins most likely evolved to evade immune recognition and to support persistent infection in the natural host, where these viruses are frequently apathogenic.
Collapse
Affiliation(s)
- Armin Ensser
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institut für Klinische und Molekulare Virologie, Schlossgarten 4, 91054 Erlangen, Germany
| |
Collapse
|
31
|
Albrecht JC, Müller-Fleckenstein I, Schmidt M, Fleckenstein B, Biesinger B. Tyrosine phosphorylation of the Tio oncoprotein is essential for transformation of primary human T cells. J Virol 2005; 79:10507-13. [PMID: 16051843 PMCID: PMC1182665 DOI: 10.1128/jvi.79.16.10507-10513.2005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human T cells are transformed to antigen-independent permanent growth in vitro upon infection with herpesvirus saimiri subgroup C strains. The viral oncoproteins required for this process, StpC and Tip, could be replaced by Tio, the oncoprotein of herpesvirus ateles. Here we demonstrate that proliferation of lymphocytes transformed with Tio-recombinant herpesvirus saimiri required the activity of Src family kinases. Src kinases had previously been identified as interaction partners of Tio. This interaction was now shown to be independent of any of the four tyrosine residues of Tio but to be dependent on an SH3-binding motif. Mutations within this motif abrogated the transforming capabilities of Tio-recombinant herpesvirus saimiri. Furthermore, kinase interaction resulted in the phosphorylation of Tio on a single tyrosine residue at position 136. Mutation of this residue in the viral context revealed that this phosphorylation site, but none of the other tyrosine residues, was required for T-cell transformation. These data indicate that the interaction of Tio with a Src kinase is essential for both the initiation and the maintenance of T-cell transformation by recombinant herpesvirus saimiri. The requirement for the tyrosine phosphorylation site at position 136 suggests a role for Tio beyond simple deregulation of the kinase.
Collapse
Affiliation(s)
- Jens-Christian Albrecht
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schlossgarten 4, D-91054 Erlangen, Germany.
| | | | | | | | | |
Collapse
|
32
|
Heck E, Lengenfelder D, Schmidt M, Müller-Fleckenstein I, Fleckenstein B, Biesinger B, Ensser A. T-cell growth transformation by herpesvirus saimiri is independent of STAT3 activation. J Virol 2005; 79:5713-20. [PMID: 15827186 PMCID: PMC1082769 DOI: 10.1128/jvi.79.9.5713-5720.2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpesvirus saimiri (saimirine herpesvirus 2) (HVS), a T-lymphotropic tumor virus, induces lymphoproliferative disease in several species of New World primates. In addition, strains of HVS subgroup C are able to transform T cells of Old World primates, including humans, to permanently growing T-cell lines. In concert with the Stp oncoprotein, the tyrosine kinase-interacting protein (Tip) of HVS C488 is required for T-cell transformation in vitro and lymphoma induction in vivo. Tip was previously shown to interact with the protein tyrosine kinase Lck. Constitutive activation of signal transducers and activators of transcription (STATs) has been associated with oncogenesis and has also been detected in HVS-transformed T-cell lines. Furthermore, Tip contains a putative consensus YXPQ binding motif for the SH2 (src homology 2) domains of STAT1 and STAT3. Tip tyrosine phosphorylation at this site was required for binding of STATs and induction of STAT-dependent transcription. Here we sought to address the relevance of STAT activation for transformation of human T cells by introducing a tyrosine-to-phenylalanine mutation in the YXPQ motif of Tip of HVS C488. Unexpectedly, the recombinant virus was still able to transform human T lymphocytes, but it had lost its capability to activate STAT3 as well as STAT1. This demonstrates that growth transformation by HVS is independent of STAT3 activation.
Collapse
Affiliation(s)
- Elke Heck
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Schlossgarten 4, D-91054 Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Signal disruption enables escape. Nat Rev Immunol 2004. [DOI: 10.1038/nri1476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|