1
|
Nalwoga A, Roshan R, Moore K, Marshall V, Miley W, Labo N, Nakibuule M, Cose S, Rochford R, Newton R, Whitby D. Kaposi's sarcoma-associated herpesvirus T cell responses in HIV seronegative individuals from rural Uganda. Nat Commun 2021; 12:7323. [PMID: 34916520 PMCID: PMC8677732 DOI: 10.1038/s41467-021-27623-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/02/2021] [Indexed: 11/09/2022] Open
Abstract
T cell responses to Kaposi's sarcoma-associated herpesvirus (KSHV) are likely essential in the control of KSHV infection and protection from associated disease, but remain poorly characterised. KSHV prevalence in rural Uganda is high at >90%. Here we investigate IFN- γ T cell responses to the KSHV proteome in HIV-negative individuals from a rural Ugandan population. We use an ex-vivo IFN- γ ELISpot assay with overlapping peptide pools spanning 83 KSHV open reading frames (ORF) on peripheral blood mononuclear cells (PBMC) from 116 individuals. KSHV-specific T cell IFN- γ responses are of low intensity and heterogeneous, with no evidence of immune dominance; by contrast, IFN- γ responses to Epstein-Barr virus, Cytomegalovirus and influenza peptides are frequent and intense. Individuals with KSHV DNA in PBMC have higher IFN- γ responses to ORF73 (p = 0.02) and lower responses to K8.1 (p = 0.004) when compared with those without KSHV DNA. In summary, we demonstrate low intensity, heterogeneous T cell responses to KSHV in immune-competent individuals.
Collapse
Affiliation(s)
- Angela Nalwoga
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda.
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| | - Romin Roshan
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kyle Moore
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Vickie Marshall
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Wendell Miley
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nazzarena Labo
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Stephen Cose
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, UK
| | - Rosemary Rochford
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | | | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
2
|
Kugasia IAR, Kumar A, Khatri A, Saeed F, Islam H, Epelbaum O. Primary effusion lymphoma of the pleural space: Report of a rare complication of cardiac transplant with review of the literature. Transpl Infect Dis 2018; 21:e13005. [PMID: 30276937 DOI: 10.1111/tid.13005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/01/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022]
Abstract
Primary effusion lymphoma (PEL) is a rare mature B-cell non-Hodgkin's lymphoma arising in body cavities and presenting with effusions. It has been described predominantly in patients with impaired immunity from the acquired immunodeficiency syndrome and is associated with the Human Herpesvirus-8 (HHV-8). Seldom has PEL been diagnosed in persons negative for the human immunodeficiency virus (HIV), and in such cases it has occurred primarily in the setting of posttransplant immunosuppression. We report an instructive case of a Caribbean-American HIV-negative orthotopic heart transplant recipient with a history of HHV-8-associated Kaposi's sarcoma who developed HHV-8 viremia and PEL of the pleural space early in the posttransplant course. This case highlights the importance of considering PEL in the differential diagnosis of a new pleural effusion in a transplant recipient at risk for HHV-8-associated disease.
Collapse
Affiliation(s)
- Irfan Ali R Kugasia
- Division of Pulmonary, Critical Care, and Sleep Medicine, Westchester Medical Center, Valhalla, New York
| | - Arun Kumar
- Department of Internal Medicine, Westchester Medical Center, Valhalla, New York
| | - Akshay Khatri
- Department of Internal Medicine, Westchester Medical Center, Valhalla, New York
| | - Faisal Saeed
- Department of Pathology, Westchester Medical Center, Valhalla, New York
| | - Humayun Islam
- Department of Pathology, Westchester Medical Center, Valhalla, New York
| | - Oleg Epelbaum
- Division of Pulmonary, Critical Care, and Sleep Medicine, Westchester Medical Center, Valhalla, New York
| |
Collapse
|
3
|
Yu L, Tu M, Cortes J, Xu-Monette ZY, Miranda RN, Zhang J, Orlowski RZ, Neelapu S, Boddu PC, Akosile MA, Uldrick TS, Yarchoan R, Medeiros LJ, Li Y, Fajgenbaum DC, Young KH. Clinical and pathological characteristics of HIV- and HHV-8-negative Castleman disease. Blood 2017; 129:1658-1668. [PMID: 28100459 PMCID: PMC5364343 DOI: 10.1182/blood-2016-11-748855] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/11/2017] [Indexed: 02/06/2023] Open
Abstract
Castleman disease (CD) comprises 3 poorly understood lymphoproliferative variants sharing several common histopathological features. Unicentric CD (UCD) is localized to a single region of lymph nodes. Multicentric CD (MCD) manifests with systemic inflammatory symptoms and organ dysfunction due to cytokine dysregulation and involves multiple lymph node regions. Human herpesvirus 8 (HHV-8) causes MCD (HHV-8-associated MCD) in immunocompromised individuals, such as HIV-infected patients. However, >50% of MCD cases are HIV and HHV-8 negative (defined as idiopathic [iMCD]). The clinical and biological behavior of CD remains poorly elucidated. Here, we analyzed the clinicopathologic features of 74 patients (43 with UCD and 31 with iMCD) and therapeutic response of 96 patients (43 with UCD and 53 with iMCD) with HIV-/HHV-8-negative CD compared with 51 HIV-/HHV-8-positive patients. Systemic inflammatory symptoms and elevated inflammatory factors were more common in iMCD patients than UCD patients. Abnormal bone marrow features were more frequent in iMCD (77.0%) than UCD (45%); the most frequent was plasmacytosis, which was seen in 3% to 30.4% of marrow cells. In the lymph nodes, higher numbers of CD3+ lymphocytes (median, 58.88 ± 20.57) and lower frequency of CD19+/CD5+ (median, 5.88 ± 6.52) were observed in iMCD patients compared with UCD patients (median CD3+ cells, 43.19 ± 17.37; median CD19+/CD5+ cells, 17.37 ± 15.80). Complete surgical resection is a better option for patients with UCD. Siltuximab had a greater proportion of complete responses and longer progression-free survival (PFS) for iMCD than rituximab. Centricity, histopathological type, and anemia significantly impacted PFS. This study reveals that CD represents a heterogeneous group of diseases with differential immunophenotypic profiling and treatment response.
Collapse
Affiliation(s)
- Li Yu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Hematology, The Second Affiliate Hospital of Nanchang University, Nanchang, China
| | - Meifeng Tu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Peking, China
| | | | - Zijun Y Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jun Zhang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sattva Neelapu
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Thomas S Uldrick
- HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - David C Fajgenbaum
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center, Houston, TX
| |
Collapse
|
4
|
Ahmad TA, Eweida AE, El-Sayed LH. T-cell epitope mapping for the design of powerful vaccines. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.vacrep.2016.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
5
|
Mousavinezhad-Moghaddam M, Amin AA, Rafatpanah H, Rezaee SAR. A new insight into viral proteins as Immunomodulatory therapeutic agents: KSHV vOX2 a homolog of human CD200 as a potent anti-inflammatory protein. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:2-13. [PMID: 27096058 PMCID: PMC4823611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The physiologic function of the immune system is defense against infectious microbes and internal tumour cells, Therefore, need to have precise modulatory mechanisms to maintain the body homeostasis. The mammalian cellular CD200 (OX2)/CD200R interaction is one of such modulatory mechanisms in which myeloid and lymphoid cells are regulated. CD200 and CD200R molecules are membrane proteins that their immunomodulatory effects are able to suppress inflammatory responses, particularly in the privilege sites such as CNS and eyes. Kaposi's sarcoma-associated herpesvirus (KSHV), encodes a wide variety of immunoregulatory proteins which play central roles in modulating inflammatory and anti-inflammatory responses in favour of virus dissemination. One such protein is a homologue of the, encoded by open reading frame (ORF) K14 and therefore called vOX2. Based on its gene expression profile during the KSHV life cycle, it is hypothesised that vOX2 modulates host inflammatory responses. Moreover, it seems that vOX2 involves in cell adhesion and modulates innate immunity and promotes Th2 immune responses. In this review the activities of mammalian CD200 and KSHV CD200 in cell adhesion and immune system modulation are reviewed in the context of potential therapeutic agents.
Collapse
Affiliation(s)
| | - Abbas Ali Amin
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Abdol Rahim Rezaee
- Inflammation and Inflammatory Diseases Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Corresponding author: Seyed Abodol Rahim Rezaee. Inflammation and Inflammatory Diseases Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. Tel: +98-51-38012768; Fax: +98-51-38436626;
| |
Collapse
|
6
|
Lee HR, Brulois K, Wong L, Jung JU. Modulation of Immune System by Kaposi's Sarcoma-Associated Herpesvirus: Lessons from Viral Evasion Strategies. Front Microbiol 2012; 3:44. [PMID: 22403573 PMCID: PMC3293256 DOI: 10.3389/fmicb.2012.00044] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Accepted: 01/27/2012] [Indexed: 12/14/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV), a member of the herpesvirus family, has evolved to establish a long-term, latent infection of cells such that while they carry the viral genome gene expression is highly restricted. Latency is a state of cryptic viral infection associated with genomic persistence in their host and this hallmark of KSHV infection leads to several clinical-epidemiological diseases such as KS, a plasmablastic variant of multicentric Castleman's disease, and primary effusion lymphoma upon immune suppression of infected hosts. In order to sustain efficient life-long persistency as well as their life cycle, KSHV dedicates a large portion of its genome to encode immunomodulatory proteins that antagonize its host's immune system. In this review, we will describe our current knowledge of the immune evasion strategies employed by KSHV at distinct stages of its viral life cycle to control the host's immune system.
Collapse
Affiliation(s)
- Hye-Ra Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | | | | | | |
Collapse
|
7
|
Tian J, Zeng G, Pang X, Liang M, Zhou J, Fang D, Liu Y, Li D, Jiang L. Identification and immunogenicity of two new HLA-A*0201-restricted CD8+ T-cell epitopes on dengue NS1 protein. Int Immunol 2012; 24:207-18. [PMID: 22298881 DOI: 10.1093/intimm/dxr115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Immunopathogenesis of dengue virus (DEN) infection remains poorly studied. Identification and characterization of human CD8(+) T-cell epitopes on DEN are necessary for a better understanding of the immunopathogenesis of dengue infection and would facilitate the development of immunotherapy and vaccines to protect from dengue infection. Here, we identified two new HLA-A*0201-restricted CD8(+) T-cell epitopes, DEN-4 NS1(990)(-998) and DEN-4 NS1(997)(-1005) that are conserved in three or four major DEN serotypes, respectively. Unexpectedly, we found that immunization of HLA-A*0201 transgenic mice with DEN-4 NS1(990)(-998) or DEN-4 NS1(997)(-1005) epitope peptide induced de novo synthesis of tumor necrosis factor (TNF)-α and IFN-γ, two important pro-inflammatory molecules that are hard to be detected directly without in vitro antigenic re-stimulation. Importantly, we demonstrated that CD8(+) T cells specifically activated by DEN-4 NS1(990)(-998) or DEN-4 NS1(997)(-1005) epitope peptide induced de novo synthesis of perforin. Furthermore, we observed that DEN-4 NS1(990)(-998) or DEN-4 NS1(997)(-1005)-specific CD8(+) T cells capable of producing large amounts of perforin, TNF-α and IFN-γ preferentially displayed CD27(+)CD45RA(-), but not CD27(-)CD45RA(+), phenotypes. This study, therefore, suggested the importance of synergistic effects of pro-inflammatory cytokines and cytotoxic molecules which were produced by dengue-specific CD8(+) T cells in immunopathogenesis or anti-dengue immunity during dengue infection.
Collapse
Affiliation(s)
- Jiang Tian
- Key Laboratory for Tropic Diseases Control, Ministry of Education of China, Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Robey RC, Mletzko S, Bower M, Meys R, Boffito M, Nelson M, Bunker CB, Gotch FM. Ex-vivo recognition of late-lytic CD8 epitopes specific for Kaposi's sarcoma-associated herpesvirus (KSHV) by HIV/KSHV-coinfected individuals. Viral Immunol 2011; 24:211-20. [PMID: 21668362 DOI: 10.1089/vim.2010.0133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS), the most common cancer in individuals with untreated HIV/AIDS. Host control of KSHV infection and KS oncogenesis by CD8 T cells remains underexplored. Although KSHV CD8 epitopes have been identified, the responses they elicit are weak and little is known about their relative importance. We sought to make a direct comparison of the recognition of a selection of the best-described known epitopes by a cohort of KSHV-seropositive, HIV-co-infected individuals, in order to assess the relative dominance of these epitopes. We further sought to identify novel epitopes from within a candidate immunogenic protein encoded by KSHV ORF28. MHC binding and denaturation assays identified putative novel A*0201-restricted epitopes from within the late-lytic glycoprotein ORF28. Recognition of these candidate epitopes was tested in a cohort of KSHV-seropositive, HIV-1-seropositive, A*0201-positive individuals by ex vivo ELISPOT, and compared with recognition of nine previously described epitopes. One novel late-lytic epitope from ORF28 was recognized by 7.1% of individuals, and was used for further investigation of KSHV-specific T cells using multimer technology. One known late-lytic epitope from the glycoprotein-encoding K8.1 was recognized by 71.4% of individuals, and represented an immunodominant KSHV epitope, but was too hydrophobic for multimer synthesis. This study identifies two KSHV CD8 epitopes derived from late-lytic antigens that are recognized by KSHV-seropositive, HIV co-infected individuals, and will be useful in future immunological studies into the CD8 response against KSHV in similar patient cohorts.
Collapse
Affiliation(s)
- Rebecca C Robey
- Department of Immunology, Imperial College London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Matthews NC, Goodier MR, Robey RC, Bower M, Gotch FM. Killing of Kaposi's sarcoma-associated herpesvirus-infected fibroblasts during latent infection by activated natural killer cells. Eur J Immunol 2011; 41:1958-68. [PMID: 21509779 PMCID: PMC3485667 DOI: 10.1002/eji.201040661] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 03/22/2011] [Accepted: 04/13/2011] [Indexed: 11/12/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) establishes life-long infection by evading clearance by the host immune system. In de novo infection and lytic replication, KSHV escapes cytotoxic T cells and NK cells through downregulation of MHC class-I and ICAM-1 molecules and associated antigens involved in forming and sustaining the immunological synapse. However, the efficacy of such mechanisms in the context of the predominantly latent KSHV infection reported in Kaposi's sarcoma (KS) lesions is unclear. Using primary dermal fibroblasts in a novel in vitro model of chronic latent KSHV infection, we generated target cells with viral loads similar to those in spindle cells extracted from KS lesions. We show that latently KSHV-infected fibroblasts had normal levels of MHC-class I, ICAM-1, HLA-E and NKG2D ligand expression, were resistant to NK-cell natural cytotoxicity and were highly susceptible to killing by cytokine-activated immunocompetent NK cells. KSHV-infected fibroblasts expressed normal levels of IFN-γR1 and responded to exogenous IFN-γ by upregulating MHC class I, ICAM-1 and HLA-E and resisting activated NK-cell killing. These data demonstrate that physiologically relevant levels of latent KSHV infection in primary cells cause limited activation of resting NK cells and confer little specific resistance to control by activated NK cells.
Collapse
Affiliation(s)
- Nick C Matthews
- Department of Immunology, Chelsea and Westminster Hospital, Imperial College London, London, UK.
| | | | | | | | | |
Collapse
|
10
|
The T-Cell Immune Response against Kaposi's Sarcoma-Associated Herpesvirus. Adv Virol 2011; 2010:340356. [PMID: 22331985 PMCID: PMC3275983 DOI: 10.1155/2010/340356] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 12/20/2010] [Indexed: 12/13/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the aetiological agent of Kaposi's sarcoma (KS), the most frequently arising malignancy in individuals with untreated HIV/AIDS. There are several lines of evidence to indicate that Kaposi's sarcoma oncogenesis is associated with loss of T-cell-mediated control of KSHV-infected cells. KSHV can establish life-long asymptomatic infection in immune-competent individuals. However, when T-cell immune control declines, for example, through AIDS or treatment with immunosuppressive drugs, both the prevalence of KSHV infection and the incidence of KS in KSHV carriers dramatically increase. Moreover, a dramatic and spontaneous improvement in KS is frequently seen when immunity is restored, for example, through antiretroviral therapy or the cessation of iatrogenic drugs. In this paper we describe the current state of knowledge on the T-cell immune responses against KSHV.
Collapse
|
11
|
Lee HR, Lee S, Chaudhary PM, Gill P, Jung JU. Immune evasion by Kaposi's sarcoma-associated herpesvirus. Future Microbiol 2011; 5:1349-65. [PMID: 20860481 DOI: 10.2217/fmb.10.105] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Persistent viral infections are often associated with serious diseases, primarily by altering functions of the host immune system. The hallmark of Kaposi's sarcoma-associated herpesvirus (KSHV) infection is the establishment of a life-long persistent infection, which leads to several clinical, epidemiological and infectious diseases, such as Kaposi's sarcoma, a plasmablastic variant of multicentric Castleman's disease, and primary effusion lymphoma. To sustain an efficient life-long persistency, KSHV dedicates a large portion of its genome to encoding immunomodulatory proteins that antagonize the immune system of its host. In this article, we highlight the strategies KSHV uses to evade, escape and survive its battle against the host's immune system.
Collapse
Affiliation(s)
- Hye-Ra Lee
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | |
Collapse
|
12
|
Dittmar T, Zänker KS. Horizontal gene transfers with or without cell fusions in all categories of the living matter. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 714:5-89. [PMID: 21506007 PMCID: PMC7120942 DOI: 10.1007/978-94-007-0782-5_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article reviews the history of widespread exchanges of genetic segments initiated over 3 billion years ago, to be part of their life style, by sphero-protoplastic cells, the ancestors of archaea, prokaryota, and eukaryota. These primordial cells shared a hostile anaerobic and overheated environment and competed for survival. "Coexist with, or subdue and conquer, expropriate its most useful possessions, or symbiose with it, your competitor" remain cellular life's basic rules. This author emphasizes the role of viruses, both in mediating cell fusions, such as the formation of the first eukaryotic cell(s) from a united crenarchaeon and prokaryota, and the transfer of host cell genes integrated into viral (phages) genomes. After rising above the Darwinian threshold, rigid rules of speciation and vertical inheritance in the three domains of life were established, but horizontal gene transfers with or without cell fusions were never abolished. The author proves with extensive, yet highly selective documentation, that not only unicellular microorganisms, but the most complex multicellular entities of the highest ranks resort to, and practice, cell fusions, and donate and accept horizontally (laterally) transferred genes. Cell fusions and horizontally exchanged genetic materials remain the fundamental attributes and inherent characteristics of the living matter, whether occurring accidentally or sought after intentionally. These events occur to cells stagnating for some 3 milliard years at a lower yet amazingly sophisticated level of evolution, and to cells achieving the highest degree of differentiation, and thus functioning in dependence on the support of a most advanced multicellular host, like those of the human brain. No living cell is completely exempt from gene drains or gene insertions.
Collapse
Affiliation(s)
- Thomas Dittmar
- Inst. Immunologie, Universität Witten/Herdecke, Stockumer Str. 10, Witten, 58448 Germany
| | - Kurt S. Zänker
- Institute of Immunologie, University of Witten/Herdecke, Stockumer Str. 10, Witten, 58448 Germany
| |
Collapse
|
13
|
Monofunctional and polyfunctional CD8+ T cell responses to human herpesvirus 8 lytic and latency proteins. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1507-16. [PMID: 20719985 DOI: 10.1128/cvi.00189-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Human herpesvirus 8 (HHV-8) is the etiological agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. It is postulated that CD8(+) T cell responses play an important role in controlling HHV-8 infection and preventing development of disease. In this study, we investigated monofunctional and polyfunctional CD8(+) T cell responses to HHV-8 lytic proteins gB (glycoprotein B) and K8.1 and latency proteins LANA-1 (latency-associated nuclear antigen-1) and K12. On the basis of our previous findings that dendritic cells (DC) reveal major histocompatibility complex (MHC) class I epitopes in gB, we used a DC-based system to identify 2 novel epitopes in gB, 2 in K8.1, 5 in LANA-1, and 1 in K12. These new HHV-8 epitopes activated monofunctional and polyfunctional CD8(+) T cells that produced various combinations of gamma interferon, interleukin 2, tumor necrosis factor alpha, macrophage inhibitory protein 1β, and cytotoxic degranulation marker CD107a in healthy HHV-8-seropositive individuals. We were also able to detect HHV-8-specific CD8(+) T cells in peripheral blood samples using HLA A*0201 pentamer complexes for one gB epitope, one K8.1 epitope, two LANA-1 epitopes, and one K12 epitope. These immunogenic regions of viral lytic and latency proteins could be important in T cell control of HHV-8 infection.
Collapse
|
14
|
Lee Y, Ferrari G, Lee SC. Estimating design space available for polyepitopes through consideration of major histocompatibility complex binding motifs. Biomed Microdevices 2010; 12:207-22. [PMID: 20033850 DOI: 10.1007/s10544-009-9376-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Major histocompatibility complex (MHC ) epitope presentation is needed for robust adaptive immune responses. Core peptide binding motifs for class I and class II MHC are 8-10 amino acids long, containing two or more "anchor" residues. These binding motifs define epitope anchor amino acid content and spacing, and knowledge of them has facilitated emergence of polyepitope vaccines. However, polyepitopes can exhibit "junctional epitopes" (neoepitopes interfering with vaccine function) resulting from juxtaposition of authentic epitopes. We have developed an algorithm for consideration of polyepitope sequence in light of MHC motifs to exhaustively identify all junctional-free polyepitope designs for any given set of authentic epitopes, and in so doing discovered that the number of such variants of any given polyepitope can be astronomically high. Our approach designs polyepitopes of any length, considers multiple MHC class I or class II motifs simultaneously and can be adapted to design variants of existing proteins with pre-selected epitope contents. We have also implemented the algorithm as a computer-based tool (CANVAC II), which we make available to interested parties. The vast diversity of junctional-free polyepitopes suggests that the number of potential T-helper epitope free protein variants may also be large, which may have implications for discovery of bioactive but non-immunogenic therapeutics.
Collapse
Affiliation(s)
- Yvonne Lee
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
15
|
Changes in T-Cell Responses Against Human Herpesvirus-8 Correlate with the Disease Course of Iatrogenic Kaposi's Sarcoma in a Patient with Undifferentiated Arthritis. Semin Arthritis Rheum 2009; 39:170-5. [DOI: 10.1016/j.semarthrit.2008.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 05/14/2008] [Accepted: 05/24/2008] [Indexed: 11/20/2022]
|
16
|
Arredouani MS, Lu B, Bhasin M, Eljanne M, Yue W, Mosquera JM, Bubley GJ, Li V, Rubin MA, Libermann TA, Sanda MG. Identification of the transcription factor single-minded homologue 2 as a potential biomarker and immunotherapy target in prostate cancer. Clin Cancer Res 2009; 15:5794-802. [PMID: 19737960 DOI: 10.1158/1078-0432.ccr-09-0911] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Identification of novel biomarkers and immunotherapy targets for prostate cancer (PCa) is crucial to better diagnosis and therapy. We sought to identify novel PCa tumor-associated antigens (TAA) that are expressed in PCa, absent in nonprostate human tissue, and immunogenic for immune responses restricted by human HLA. EXPERIMENTAL DESIGN AND RESULTS Using microarray analysis of normal and cancerous human prostate tissues, we identified 1,063 genes overexpressed in PCa. After validating 195 transcripts in publicly available array data sets, we interrogated expression of these TAAs in normal human tissues to identify genes that are not expressed at detectable levels in normal, nonprostate adult human tissue. We identified 23 PCa TAA candidates. Real-time PCR confirmed that 15 of these genes were overexpressed in PCa (P< 0.05 for each). The most frequently overexpressed gene, single-minded homologue 2 (SIM2), was selected for further evaluation as a potential target for immunotherapy. ELISA assay revealed that a fraction of PCa patients exhibited immune responsiveness to SIM2 as evidenced by the presence of autoantibodies to SIM2 in their sera. We next showed binding of putative HLA-A2.1-restricted SIM2 epitopes to human A2.1, and immunization of transgenic HLA-A2.1 mice showed induction of SIM2-specific CTL responses in vivo. CONCLUSIONS Our findings that SIM2 is selectively expressed in PCa, that human HLA-A2.1-restricted SIM2 epitopes induce specific T cells in vivo, and that anti-SIM2 antibodies are detectable in PCa patients' sera implicate SIM2 as a PCa-associated antigen that is a suitable potential target for PCa immunotherapy.
Collapse
Affiliation(s)
- Mohamed S Arredouani
- Division of Urology, Department of Surgery, Genomics Center, and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Robey RC, Lagos D, Gratrix F, Henderson S, Matthews NC, Vart RJ, Bower M, Boshoff C, Gotch FM. The CD8 and CD4 T-cell response against Kaposi's sarcoma-associated herpesvirus is skewed towards early and late lytic antigens. PLoS One 2009; 4:e5890. [PMID: 19536280 PMCID: PMC2691989 DOI: 10.1371/journal.pone.0005890] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 04/30/2009] [Indexed: 01/06/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is causally related to Kaposi's sarcoma (KS), the most common malignancy in untreated individuals with HIV/AIDS. The adaptive T-cell immune response against KSHV has not been fully characterized. To achieve a better understanding of the antigenic repertoire of the CD8 and CD4 T-cell responses against KSHV, we constructed a library of lentiviral expression vectors each coding for one of 31 individual KSHV open reading frames (ORFs). We used these to transduce monocyte-derived dendritic cells (moDCs) isolated from 14 KSHV-seropositive (12 HIV-positive) and 7 KSHV-seronegative (4 HIV-positive) individuals. moDCs were transduced with up to 3 KSHV ORFs simultaneously (ORFs grouped according to their expression during the viral life cycle). Transduced moDCs naturally process the KSHV genes and present the resulting antigens in the context of MHC class I and II. Transduced moDCs were cultured with purified autologous T cells and the CD8 and CD4 T-cell proliferative responses to each KSHV ORF (or group) was assessed using a CFSE dye-based assay. Two pools of early lytic KSHV genes ([ORF8/ORF49/ORF61] and [ORF59/ORF65/K4.1]) were frequently-recognized targets of both CD8 and CD4 T cells from KSHV seropositive individuals. One pool of late lytic KSHV genes ([ORF28/ORF36/ORF37]) was a frequently-recognized CD8 target and another pool of late genes ([ORF33/K1/K8.1]) was a frequently-recognized CD4 target. We report that both the CD8 and CD4 T-cell responses against KSHV are skewed towards genes expressed in the early and late phases of the viral lytic cycle, and identify some previously unknown targets of these responses. This knowledge will be important to future immunological investigations into KSHV and may eventually lead to the development of better immunotherapies for KSHV-related diseases.
Collapse
Affiliation(s)
- Rebecca C. Robey
- Department of Immunology, Imperial College London, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Dimitrios Lagos
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Fiona Gratrix
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Stephen Henderson
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Nick C. Matthews
- Department of Immunology, Imperial College London, London, United Kingdom
| | - Richard J. Vart
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Mark Bower
- Department of Immunology, Imperial College London, London, United Kingdom
| | - Chris Boshoff
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Frances M. Gotch
- Department of Immunology, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
18
|
CD8+ T cell immunity to Epstein-Barr virus and Kaposi's sarcoma-associated herpes virus. Semin Cancer Biol 2008; 18:416-22. [PMID: 19007888 DOI: 10.1016/j.semcancer.2008.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 10/16/2008] [Indexed: 12/27/2022]
Abstract
Gammaherpesviruses are agents which have evolved to persist within the lymphoid system and many have oncogenic potential; studying gammaherpesvirus infections therefore has the potential to reveal much about the workings of the immune system and the control over viral oncogenesis. The lymphocryptovirus Epstein-Barr virus (EBV) and the rhadinovirus Kaposi's sarcoma-associated herpesvirus (KSHV, also known as human herpesvirus 8) are the two human gammaherpesviruses. Analysis of the T cell response to EBV has guided understanding of immunity to infection and disease caused by this virus, as well as directed the development of vaccination and therapeutic interventions in EBV-associated disease. Less is known about the T cell response to KSHV and its exact role in controlling virus infection and disease. Here we discuss the CD8+ T cell response to these two gammaherpesviruses.
Collapse
|
19
|
Changes in the Immune Responses Against Human Herpesvirus-8 in the Disease Course of Posttransplant Kaposi Sarcoma. Transplantation 2008; 86:738-44. [DOI: 10.1097/tp.0b013e318184112c] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Barcy S, De Rosa SC, Vieira J, Diem K, Ikoma M, Casper C, Corey L. Gamma delta+ T cells involvement in viral immune control of chronic human herpesvirus 8 infection. THE JOURNAL OF IMMUNOLOGY 2008; 180:3417-25. [PMID: 18292568 DOI: 10.4049/jimmunol.180.5.3417] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Little is known about what effector populations are associated with the control of human herpesvirus 8 (HHV-8) infection in vivo. We compared T lymphocyte subsets among HIV-HHV-8+ and HIV-HHV-8- infected human individuals. alphabeta+ T cells from HHV-8-infected individuals displayed a significantly higher percentage of differentiated effector cells among both CD4+ and CD8+ T cell subsets. HHV-8 infection was associated with significant expansion of gammadelta+ Vdelta1 T cells expressing a differentiated effector cell phenotype in peripheral blood. In vitro stimulation of PBMC from HHV-8-infected individuals with either infectious viral particles or different HHV-8 viral proteins resulted in gammadelta Vdelta1 T cell activation. In addition, gammadelta Vdelta1 T cells displayed a strong reactivity against HHV-8-infected cell lines and prevented the release of infectious viral particles following the induction of lyric replication. These data indicate that gammadelta T cells play a role in both innate and adaptive T cell responses against HHV-8 in immunocompetent individuals.
Collapse
Affiliation(s)
- Serge Barcy
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98109, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Multicentric Castleman disease is associated with polyfunctional effector memory HHV-8-specific CD8+ T cells. Blood 2007; 111:1387-95. [PMID: 18000163 DOI: 10.1182/blood-2007-03-080648] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Multicentric Castleman disease (MCD) is a devastating human herpesvirus 8 (HHV-8)-related lymphoproliferative disorder that occurs in immunocompromised persons. To determine the role of immune responses in MCD, we studied the frequency, antigenic repertoire, differentiation, and functional profile of HHV-8-specific CD8(+) T cells in MCD patients and in human immunodeficiency virus-coinfected asymptomatic HHV-8 carriers (AC). Screening CD8(+) T-cell responses with ELISpot interferon-gamma (IFN-gamma) assays using 56 peptides on 6 latent and lytic HHV-8 proteins showed that MCD and AC patients had responses of similar magnitude and antigenic repertoire and identified a new 10-mer human leukocyte antigen B7 CD8 epitope in K15. Intracellular IFN-gamma staining showed significantly more CD45RA(-)CCR7(-)CD27(-) CD8(+)IFN-gamma(+) cells (late phenotype) and significantly fewer CCR7(-)CD27(+)CD45RA(-) cells (early and intermediate phenotype) in MCD than in AC patients. This phenotypic shift was not found for Epstein-Barr virus-specific CD8(+) T cells tested as controls. HHV-8 viral loads were negatively correlated with early and intermediate effector memory cells. HHV-8-specific T cells were polyfunctional (secretion of IFN-gamma, tumor necrosis factor-alpha, macrophage inflammatory protein-1beta, and/or CD107a) in both MCD and AC patients. In conclusion, MCD is not associated with a lack of HHV-8-specific CD8(+) T cells or limitation of their functional profile. Their differentiation increases with HHV-8 viral load. These results offer new insight into the pathophysiology of MCD.
Collapse
|
22
|
Stebbing J, Sanitt A, Teague A, Powles T, Nelson M, Gazzard B, Bower M. Prognostic Significance of Immune Subset Measurement in Individuals With AIDS-Associated Kaposi's Sarcoma. J Clin Oncol 2007; 25:2230-5. [PMID: 17470847 DOI: 10.1200/jco.2007.10.7219] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Purpose A prognostic index for AIDS-associated Kaposi's sarcoma (KS) diagnosed in the era of highly active antiretroviral therapy (HAART) was based on routine clinical and laboratory characteristics. Because immune subset measurement is often performed in HIV-positive individuals, we examined whether these were predictive of mortality independently of the prognostic index, or could predict time to progression of KS. Patients and Methods We performed univariate and multivariate Cox regression analyses on a data set of 326 individuals with AIDS-associated KS to identify immune subset covariates predictive of overall survival and time to progression. Adaptive (CD8 T cell and CD19 B cell) and innate (CD16/56 natural-killer cell) immune parameters were studied by flow cytometry. Results In univariate analyses, all three immune subsets had significant effects on overall survival (P < .025). In multivariate analyses including the prognostic index, only CD8 counts remained significant (P = .026), although its effect on the overall prognostic index is small. An increase of 100 cells/mm3 in the CD8 count confers a 5% improvement in overall survival. Individuals with a higher CD8 count did not have an increased time to progression. Patients who were already on HAART at the time of KS diagnosis did not have a shorter time to progression than those who were antiretroviral naïve at KS diagnosis. Conclusion The CD8 count appears to provide independent prognostic information in individuals with AIDS-associated KS. Measurement of the CD8 count is clinically useful in patients with KS.
Collapse
Affiliation(s)
- Justin Stebbing
- Imperial College of Science, Medicine, and Technology, Department of Oncology, The Chelsea and Westminster Hospital, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
23
|
Sundar K, Boesen A, Coico R. Computational prediction and identification of HLA-A2.1-specific Ebola virus CTL epitopes. Virology 2006; 360:257-63. [PMID: 17123567 DOI: 10.1016/j.virol.2006.09.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Revised: 07/05/2006] [Accepted: 09/27/2006] [Indexed: 11/24/2022]
Abstract
Ebola virus (EBOV) is known to cause a severe hemorrhagic fever resulting in high mortality. Although the precise host defense mechanism(s) that afford protection against EBOV is not completely understood, T cell-mediated immune responses is believed to play a pivotal role in controlling virus replication and EBOV infection. There have been no reports on mapping of MHC Class I-binding CTL epitopes for EBOV till to date. In this study, we identified five HLA-A2-binding 9-mer peptides of EBOV nucleoprotein (NP) using computer-assisted algorithm. The peptides were synthesized and examined for their ability to bind to MHC class I molecules using a flow cytometry based MHC stabilization assay. Three of the EBOV-NP peptides tested (FLSFASLFL, RLMRTNFLI and KLTEAITAA) stabilized HLA-A2. The ability of the HLA-A2-binding EBOV-NP peptides to generate peptide-specific CTLs was evaluated in HLA-A2.1 transgenic mice. Epitope-specific CTL responses were confirmed by cytotoxic assays against peptide-pulsed target cells and interferon-gamma ELISPOT assay. Each of the EBOV-NP peptides induced CTL responses in HLA-A2-transgenic mice. Interestingly, all the three peptides were conserved in three different strains of Ebola (Zaire and Reston and Sudan). Taken together, these findings provide direct evidence for the existence of EBOV-derived NP epitopes that may be useful in the development of protective immunogens for this hemorrhagic virus.
Collapse
Affiliation(s)
- Krishnan Sundar
- Department of Microbiology and Immunology, City University of New York Medical School, New York, NY 10031, USA
| | | | | |
Collapse
|
24
|
Lambert M, Gannagé M, Karras A, Abel M, Legendre C, Kerob D, Agbalika F, Girard PM, Lebbe C, Caillat-Zucman S. Differences in the frequency and function of HHV8-specific CD8 T cells between asymptomatic HHV8 infection and Kaposi sarcoma. Blood 2006; 108:3871-80. [PMID: 16926293 DOI: 10.1182/blood-2006-03-014225] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
It is unclear how the immune response controls human herpesvirus 8 (HHV8; also known as Kaposi sarcoma-associated herpesvirus [KSHV]) replication and thereby prevents Kaposi sarcoma (KS). We compared CD8 T-cell responses to HHV8 latent (K12) and lytic (glycoprotein B, ORF6, ORF61, and ORF65) antigens in patients who spontaneously controlled the infection and in patients with posttransplantation, AIDS-related, or classical KS. We found that anti-HHV8 responses were frequent, diverse, and strongly differentiated toward an effector phenotype in patients who controlled the infection. Conversely, HHV8-specific CD8 cells were very rare in patients who progressed to KS, and were not recruited to the tumoral tissue, as visualized by in situ tetramer staining of KS biopsies. Last, HHV8-specific CD8 T cells were observed in a seronegative recipient of an HHV8infected graft who remained persistently aviremic and antibody negative, suggesting that specific cytotoxic T lymphocytes (CTLs) may provide protection from persistent HHV8 infection. These results support the crucial role of cellular immune responses in controlling HHV8 replication, in preventing malignancies in latently infected subjects, and in conferring genuine resistance to persistent infection. They may also have important implications for the design of prophylactic and therapeutic HHV8 vaccines, and for adoptive immunotherapy of KS.
Collapse
MESH Headings
- Acquired Immunodeficiency Syndrome/complications
- Acquired Immunodeficiency Syndrome/immunology
- Acquired Immunodeficiency Syndrome/pathology
- Acquired Immunodeficiency Syndrome/virology
- Aged
- Antigens, Viral, Tumor/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Female
- Herpesvirus 8, Human/immunology
- Herpesvirus Vaccines/immunology
- Herpesvirus Vaccines/therapeutic use
- Humans
- Immunotherapy, Adoptive/methods
- Male
- Middle Aged
- Neoplasm Regression, Spontaneous/immunology
- Neoplasm Regression, Spontaneous/pathology
- Sarcoma, Kaposi/immunology
- Sarcoma, Kaposi/pathology
- Sarcoma, Kaposi/therapy
- Sarcoma, Kaposi/virology
- Transplants/adverse effects
- Virus Replication/immunology
Collapse
Affiliation(s)
- Marion Lambert
- INSERM U561, Hôpital St-Vincent de Paul, 75014 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bego MG, St Jeor S. Human cytomegalovirus infection of cells of hematopoietic origin: HCMV-induced immunosuppression, immune evasion, and latency. Exp Hematol 2006; 34:555-70. [PMID: 16647557 DOI: 10.1016/j.exphem.2005.11.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Revised: 11/15/2005] [Accepted: 11/21/2005] [Indexed: 12/16/2022]
Affiliation(s)
- Mariana G Bego
- Department of Microbiology and Immunology, University of Nevada, Reno, Reno, NV 89557, USA
| | | |
Collapse
|
26
|
Boesen A, Sundar K, Coico R. Lassa fever virus peptides predicted by computational analysis induce epitope-specific cytotoxic-T-lymphocyte responses in HLA-A2.1 transgenic mice. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 12:1223-30. [PMID: 16210487 PMCID: PMC1247823 DOI: 10.1128/cdli.12.10.1223-1230.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2005] [Revised: 05/17/2005] [Accepted: 06/16/2005] [Indexed: 11/20/2022]
Abstract
Lassa fever is a hemorrhagic disease caused by Lassa fever virus (LV). Although the precise host defense mechanism(s) that affords protection against LV is not completely understood, cellular immunity mediated by cytotoxic T lymphocytes (CTLs) plays a pivotal role in controlling viral replication and LV infection. To date, there have been no reports mapping major histocompatibility complex (MHC) class I-binding CTL epitopes for LV. Using computer-assisted algorithms, we identified five HLA-A2.1-binding peptides of LV glycoprotein (GP) and two peptides from LV nucleoprotein (NP). Synthesized peptides were examined for their ability to bind to MHC class I molecules using a flow cytometric assay that measures peptide stabilization of class I. Three of the LV-GP peptides tested (LLGTFTWTL, SLYKGVYEL, and YLISIFLHL) stabilized HLA-A2. The LV-NP peptides tested failed to stabilize this HLA-A2. We then investigated the ability of the HLA-A2-binding LV-GP peptides to generate peptide-specific CTLs in HLA-A2.1 transgenic mice. Functional assays used to confirm CTL activation included gamma interferon enzyme-linked immunospot (ELISPOT) assays and intracellular cytokine staining of CD8+ T cells from peptide-primed mice. CTL assays were also performed to verify the cytolytic activity of peptide-pulsed target cells. Each of the LV-GP peptides induced CTL responses in HLA-A2-transgenic mice. MHC class I tetramers prepared using one LV-GP peptide that showed the highest cytolytic index (LLGTFTWTL) confirmed that peptide-binding CD8+ T cells were present in pooled lymphocytes harvested from peptide-primed mice. These findings provide direct evidence for the existence of LV-derived GP epitopes that may be useful in the development of protective immunogens for this hemorrhagic virus.
Collapse
Affiliation(s)
- Agnieszka Boesen
- Department of Microbiology and Immunology, City University of New York Medical School, New York, New York 10031, USA
| | | | | |
Collapse
|
27
|
Tomkowicz B, Singh SP, Lai D, Singh A, Mahalingham S, Joseph J, Srivastava S, Srinivasan A. Mutational analysis reveals an essential role for the LXXLL motif in the transformation function of the human herpesvirus-8 oncoprotein, kaposin. DNA Cell Biol 2005; 24:10-20. [PMID: 15684715 DOI: 10.1089/dna.2005.24.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Human herpesvirus-8 (HHV-8) is causally linked to Kaposi's sarcoma (KS). Sequence analysis of the genome and subsequent studies revealed several genes including kaposin, with transformation properties in cell culture. In this study, we have analyzed the requirement of Kaposin A for cellular transformation in an effort to understand its contribution towards KS pathogenesis. Comparative analysis of Kaposin with other proteins identified the LXXLL motif spanning from residues 31-35 (LVCLL). The observation that the LXXLL motif is present in nuclear receptor coactivators that mediate the interaction of coactivators with nuclear receptors has prompted us to investigate the relevance of this motif for Kaposin's function(s). Kaposin A coding sequences were cloned into a eukaryotic expression plasmid with the Flag (FL) epitope fused in-frame at the C-terminus (Kap-FL). To evaluate the role of leucine residues in the motif, site-directed mutagenesis was utilized, whereby alanine was substituted for the leucine residues (Kap-AXXAA-FL). Both Kap-FL and Kap- AXXAA-FL exhibited similar levels of expression in cells. Interestingly, the Kap-AXXAA-FL mutant failed to show transforming activity by two independent assays: anchorage-independent growth, and focus formation. Immunofluorescence (IFA) and FACS analysis indicated that Kap-FL was localized around the nucleus and at the cell surface, respectively. However, Kap-AXXAA-FL exhibited diffuse cytoplasmic staining as measured by IFA yet was still detectable on the cell surface by FACS. Ironically, both Kap-FL and Kap-AXXAAFL were able to activate the AP-1 promoter. These results support an important role for the LXXLL motif in the ability of Kaposin to induce transformation.
Collapse
MESH Headings
- Amino Acid Motifs/genetics
- Amino Acid Sequence
- Animals
- Cell Nucleus/immunology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Viral/genetics
- DNA Mutational Analysis
- Herpesvirus 8, Human/genetics
- Humans
- Leucine/genetics
- Mice
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation/genetics
- NIH 3T3 Cells
- Oncogene Proteins, Viral/chemistry
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/metabolism
- Promoter Regions, Genetic/genetics
- Sarcoma, Kaposi/genetics
- Sarcoma, Kaposi/metabolism
- Sarcoma, Kaposi/virology
- Transcription Factor AP-1/genetics
- Transfection
- Viral Proteins/chemistry
- Viral Proteins/genetics
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Brian Tomkowicz
- Department of Microbiology and Immunology, Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Caputo A, Brocca-Cofano E, Castaldello A, De Michele R, Altavilla G, Marchisio M, Gavioli R, Rolen U, Chiarantini L, Cerasi A, Dominici S, Magnani M, Cafaro A, Sparnacci K, Laus M, Tondelli L, Ensoli B. Novel biocompatible anionic polymeric microspheres for the delivery of the HIV-1 Tat protein for vaccine application. Vaccine 2004; 22:2910-24. [PMID: 15246628 DOI: 10.1016/j.vaccine.2003.12.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2003] [Accepted: 12/02/2003] [Indexed: 10/26/2022]
Abstract
Two novel classes of biocompatible core-shell anionic microspheres, composed of an inner hard insoluble core, either made of poly(styrene) (PS) or poly(methyl methacrylate) (PMMA), and a soft outer tentacular shell made of long soluble negatively charged arms derived from the steric stabilizer, hemisuccinated poly(vinyl alcohol) or Eudragit L100/55, respectively, were prepared by dispersion polymerization and characterized. Five types of these novel microspheres, two made of poly(styrene) and hemisuccinated poly(vinyl alcohol) (A4 and A7), and three made of poly(methyl methacrylate) and Eudragit L100/55 (1D, 1E, H1D), differing for chemical composition, size, and surface charge density were analyzed for the delivery of the HIV-1 Tat protein for vaccine applications. All microspheres reversibly adsorbed the native biologically active HIV-1 Tat protein preventing Tat from oxidation and maintaining its biological activity, therefore increasing the shelf-life of the Tat protein vaccine. The microspheres efficiently delivered Tat intracellularly, and were not toxic in vitro nor in mice, even after multiple administrations. These results indicate that these novel microparticles are safe and represent a promising delivery system for vaccination with Tat, as well as for other subunit vaccines, particularly when a native protein conformation is required.
Collapse
Affiliation(s)
- Antonella Caputo
- Department of Experimental and Diagnostic Medicine, Section of Microbiology, University of Ferrara, Via Luigi Borsari 46, 44100, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Henke-Gendo C, Schulz TF. Transmission and disease association of Kaposi's sarcoma-associated herpesvirus: recent developments. Curr Opin Infect Dis 2004; 17:53-7. [PMID: 15090892 DOI: 10.1097/00001432-200402000-00011] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Kaposi's sarcoma-associated herpesvirus or human herpesvirus 8, common in sub-Saharan Africa and around the Mediterranean Sea but rare in most other countries, is known to be transmitted in childhood within families in endemic regions, and through sexual contacts among high-risk groups in Western countries. Nevertheless recent developments on other modes of transmission of the virus have been made during the last years and are summarized in this review. Furthermore, recent published disease associations are discussed. RECENT FINDINGS The last year has seen research addressing the question of parenteral transmission, sexual transmission through heterosexual contact, transmission of Kaposi's sarcoma-associated herpesvirus-infected cells from organ donors to recipient, as well as the first suggestion that host genetic factors may facilitate infection in childhood. Additional clinical manifestations of infection with the virus such as primary pulmonary hypertension and germinotropic lymphoproliferative disorder have been identified. SUMMARY Evidence of Kaposi's sarcoma-associated herpesvirus transmission other than between homosexual adults and during childhood - namely transmission through heterosexual contact or injection drug use - is growing although these issues are still incompletely analysed and far away from being fully understood. Despite our increasing knowledge on transmission and disease associations of the virus, implications on the clinical management of associated diseases and public health have to be further evaluated in the coming years.
Collapse
|
30
|
Ramachandran S, Jaramillo A, Xu XC, McKane BW, Chapman WC, Mohanakumar T. HUMAN IMMUNE RESPONSES TO PORCINE ENDOGENOUS RETROVIRUS-DERIVED PEPTIDES PRESENTED NATURALLY IN THE CONTEXT OF PORCINE AND HUMAN MAJOR HISTOCOMPATIBILITY COMPLEX CLASS I MOLECULES: IMPLICATIONS IN XENOTRANSPLANTATION OF PORCINE ORGANS. Transplantation 2004; 77:1580-8. [PMID: 15239626 DOI: 10.1097/01.tp.0000122220.61309.1d] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Porcine endogenous retroviruses (PERV) have been shown to infect human cells, raising concerns regarding safety of xenotransplantation. In patients exposed to porcine tissues, no PERV infection has been observed. This study was designed to develop human CD8+ cytotoxic T lymphocytes (CTL) against PERV-derived peptides presented in the context of human leukocyte antigen (HLA) or swine leukocyte antigen (SLA) class I molecules and to define dominant epitopes contributed by PERV. METHODS Human CD8+ CTL were generated against porcine aortic endothelial cells (PAEC). Peptides presented on SLA class I molecules were acid eluted and fractionated by reverse-phase high-performance liquid chromatography. Peptide fractions that restored lysis of acid-stripped PAEC were sequenced by tandem mass spectrometry. Human CD8+ CTL were generated against PERV envelope-derived peptides and PERV-infected human cells to identify immunodominant PERV-derived epitopes. RESULTS We identified two peptides derived from retroviral transactivating regulatory protein (AHQDPLPEQP) and retroviral transcription factor (PQKPFVT) recognized by human CD8+ CTL in the context of SLA class I. Computer-assisted analysis identified nine PERV-envelope-derived 9-mer peptides with high affinity for the HLA-A2 molecule (Env-1-9). PERV-specific CD8+ CTL generated in vitro identified the immunodominant Env-5 peptide (303-311, KLFSLIQGA) and demonstrated HLA-A2-restricted cytotoxicity against PERV-infected human cells. CONCLUSIONS Our results indicate that PERV-derived peptides are presented naturally on porcine and human major histocompatibility complex class I molecules. CD8+ CTL responses elicited against dominant SLA and HLA class I-restricted PERV-derived epitopes may play an important role in xenograft rejection and in containment of PERV infection of human cells after xenotransplantation.
Collapse
Affiliation(s)
- Sabarinathan Ramachandran
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
31
|
Bourboulia D, Aldam D, Lagos D, Allen E, Williams I, Cornforth D, Copas A, Boshoff C. Short- and long-term effects of highly active antiretroviral therapy on Kaposi sarcoma-associated herpesvirus immune responses and viraemia. AIDS 2004; 18:485-93. [PMID: 15090801 DOI: 10.1097/00002030-200402200-00015] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the effect of highly active antiretroviral therapy (HAART) on Kaposi sarcoma-associated herpesvirus (KSHV) DNA load, anti-KSHV antibody responses and KSHV-specific CD8 T cell responses in HIV-infected individuals over a 2 year period. DESIGN Prospective study of 27 HIV-infected antiretroviral therapy-naive individuals, with (n = 4) and without KS (n = 23), before HAART and at 3-month intervals, during treatment with HAART. METHODS Sequential blood samples were collected for anti-KSHV antibody detection, KSHV DNA load in peripheral blood mononuclear cells (PBMC) and plasma, HIV Gag-specific and KSHV-specific CD8 T cell responses, HIV-1 plasma RNA load and CD4 and CD8 T cell counts. RESULTS KSHV DNA in PBMC and plasma became less detectable over time during HAART, in particular after 12 months. KSHV DNA was undetectable in plasma after 24 months on HAART. Anti-KSHV lytic, but not latent, antibody levels increased within 12 months of treatment. KSHV-specific CD8 T cell responses were absent prior to HAART but became detectable in some patients within 6 months of starting treatment, and continued to increase thereafter. CONCLUSIONS HAART (both protease inhibitor-based and non-nucleoside reverse transcriptase inhibitor-based antiretroviral combinations) is associated with immune reconstitution to KSHV and with undetectable KSHV viraemia. However, this restoration is apparent (in particular) only after a relatively long (> 24 months) period of treatment. These immune responses could contribute to the decreased incidence of KS during HAART, but it is unlikely to be a complete explanation for the often rapid resolution of KS when HAART is started.
Collapse
Affiliation(s)
- Dimitra Bourboulia
- Wolfson Institute for Biomedical Research and the Department of Sexually Transmitted Diseases, University College London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Tomescu C, Law WK, Kedes DH. Surface downregulation of major histocompatibility complex class I, PE-CAM, and ICAM-1 following de novo infection of endothelial cells with Kaposi's sarcoma-associated herpesvirus. J Virol 2003; 77:9669-84. [PMID: 12915579 PMCID: PMC187401 DOI: 10.1128/jvi.77.17.9669-9684.2003] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Under selective pressure from host cytotoxic T lymphocytes, many viruses have evolved to downregulate major histocompatibility complex (MHC) class I and/or T-cell costimulatory molecules from the surface of infected cells. Kaposi's sarcoma-associated herpesvirus (KSHV) encodes two proteins, MIR-1 and MIR-2, that serve this function during lytic replication. In vivo, however, KSHV exists in a predominantly latent state, with less than 5% of infected cells expressing discernible lytic gene products. Thus, mechanisms of immune evasion that depend on genes expressed only during lytic replication are unlikely to be active in most KSHV-infected cells. As a result, we searched for evidence of similar defensive strategies extant during latency, employing culture systems that strongly favor latent KSHV infection. We measured cell surface levels of immunomodulatory proteins on both primary dermal microvascular endothelial cells (pDMVEC) infected through coculture with induced primary effusion lymphoma cells and telomerase-immortalized DMVEC infected directly with cell-free virus. Employing a panel of antibodies against several endothelial cell surface proteins, we show that de novo infection with KSHV leads to the downregulation of MHC class I, CD31 (PE-CAM), and CD54 (ICAM-I) but not CD58 (LFA-3) or CD95 (Fas). Furthermore, flow cytometry with a fluorescently labeled monoclonal antibody to the latency-associated nuclear antigen (LANA) revealed that downregulation occurred predominantly on KSHV-infected (LANA-positive) cells. Although the vast majority of infected cells displayed this downregulation, less than 1% expressed either immediate-early or late lytic proteins detectable by immunofluorescence. Together, these results suggest that downregulation of immunomodulatory proteins on the surface of target cells may represent a constitutive mode of immune evasion employed by KSHV following de novo infection.
Collapse
Affiliation(s)
- Costin Tomescu
- Myles H. Thaler Center for AIDS and Human Retrovirus Research, Department of Microbiology, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | |
Collapse
|
33
|
Webb AI, Aguilar MI, Purcell AW. Optimisation of peptide-based cytotoxic T-cell determinants using non-natural amino acids. ACTA ACUST UNITED AC 2003. [DOI: 10.1007/bf02442589] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
34
|
Sette A, Fikes J. Epitope-based vaccines: an update on epitope identification, vaccine design and delivery. Curr Opin Immunol 2003; 15:461-70. [PMID: 12900280 DOI: 10.1016/s0952-7915(03)00083-9] [Citation(s) in RCA: 240] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The basic premise of the epitope-based approach to vaccine development is that, in certain cases, the responses induced by the natural immunogen are not optimal, and can be improved upon by isolation or optimization of specific components of the response. For example, immunodominance is a key factor limiting the type and breadth of adaptive immunity. Recent advances in understanding the mechanisms of immunodominance thus represent an opportunity to further develop the epitope-based approach.
Collapse
Affiliation(s)
- Alessandro Sette
- La Jolla Institute for Allergy and Immunology, San Diego, California 92121, USA.
| | | |
Collapse
|
35
|
Stebbing J, Bourboulia D, Johnson M, Henderson S, Williams I, Wilder N, Tyrer M, Youle M, Imami N, Kobu T, Kuon W, Sieper J, Gotch F, Boshoff C. Kaposi's sarcoma-associated herpesvirus cytotoxic T lymphocytes recognize and target Darwinian positively selected autologous K1 epitopes. J Virol 2003; 77:4306-14. [PMID: 12634388 PMCID: PMC150628 DOI: 10.1128/jvi.77.7.4306-4314.2003] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the infectious cause of Kaposi's sarcoma (KS) and certain lymphoproliferations particularly in the context of human immunodeficiency virus (HIV) type 1-induced immunosuppression. The introduction of effective therapies to treat HIV has led to a decline in the incidence of KS, suggesting that immune responses may play a role in controlling KSHV infection and pathogenesis. Cytotoxic-T-lymphocyte (CTL) activity against KSHV proteins has been demonstrated; however, the identification of KSHV CTL epitopes remains elusive and problematic. Although the herpesvirus genomic layout is generally conserved, KSHV encodes a unique hypervariable protein, K1, with intense biological selection pressure at specific amino acid sites. To investigate whether this variability is partly driven by cellular immunity, we designed K1 peptides that match only the unique viral sequence for every individual studied here (autologous peptides). We identified functional CTL epitopes within K1's most variable areas, and we show that a given individual responds only to autologous peptides and not to peptides from other individuals. Furthermore, these epitopes are highly conserved sequences within KSHV isolates from a specific strain but are not conserved between different strains. We conclude that CTL recognition contributes to K1, and therefore to KSHV, evolution.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigenic Variation
- Antigens, Viral/genetics
- Base Sequence
- Cytotoxicity, Immunologic
- DNA, Viral/genetics
- Epitopes/genetics
- HIV Infections/complications
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/immunology
- Herpesvirus 8, Human/pathogenicity
- Humans
- In Vitro Techniques
- Molecular Sequence Data
- Sarcoma, Kaposi/complications
- Sarcoma, Kaposi/immunology
- Sarcoma, Kaposi/virology
- Selection, Genetic
- Sequence Homology, Amino Acid
- T-Lymphocytes, Cytotoxic/immunology
- Viral Proteins/genetics
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Justin Stebbing
- Cancer Research U.K. Viral Oncology Group, The Wolfson Institute for Biomedical Research, University College London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Stebbing J, Portsmouth S, Bower M. Insights into the molecular biology and sero-epidemiology of Kaposi's sarcoma. Curr Opin Infect Dis 2003; 16:25-31. [PMID: 12821826 DOI: 10.1097/00001432-200302000-00005] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Although the incidence of Kaposi's sarcoma in established market economies has fallen since the introduction of highly active antiretroviral therapy, it remains the most common tumour in individuals with HIV infection. Kaposi's sarcoma-associated herpesvirus is the aetiological agent of Kaposi's sarcoma and its role in the subversion of cellular machinery provides an understanding of the fundamental mechanisms involved in immunobiology and carcinogenesis. The focus of this review will be to discuss articles published from August 2001 to August 2002 that provide advances in our knowledge of this process, and also to review new data concerning the transmission and identification of Kaposi's sarcoma-associated herpesvirus infection in different populations. RECENT FINDINGS We have an improved understanding of the epidemiologies of classic, endemic, post-transplant and AIDS-associated Kaposi's sarcoma. The role of specific genes in tumorigenesis has been further defined, with particular reference to the switch from latent to lytic infection and the involvement of cytokines and angiogenic factors. SUMMARY Kaposi's sarcoma-associated herpesvirus is widespread in immunosuppressed individuals and is associated with significant morbidity and mortality. The study of Kaposi's sarcoma and Kaposi's sarcoma-associated herpesvirus represents an important model of the interplay between the immune system and cancer, and may lead to the development of clinically useful therapies including vaccines.
Collapse
|