1
|
Li X, Ohler ZW, Day A, Bassel L, Grosskopf A, Afsari B, Tagawa T, Custer W, Mangusan R, Lurain K, Yarchoan R, Ziegelbauer J, Ramaswami R, Krug LT. Mapping herpesvirus-driven impacts on the cellular milieu and transcriptional profile of Kaposi sarcoma in patient-derived mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615429. [PMID: 39386738 PMCID: PMC11463583 DOI: 10.1101/2024.09.27.615429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Kaposi sarcoma (KS) is defined by aberrant angiogenesis driven by Kaposi sarcoma herpesvirus (KSHV)-infected spindle cells with endothelial characteristics. KS research is hindered by rapid loss of KSHV infection upon explant culture of tumor cells. Here, we establish patient-derived KS xenografts (PDXs) upon orthotopic implantation of cutaneous KS biopsies in immunodeficient mice. KS tumors were maintained in 27/28 PDX until experimental endpoint, up to 272 days in the first passage of recipient mice. KSHV latency associated nuclear antigen (LANA)+ endothelial cell density increased by a mean 4.3-fold in 14/15 PDX analyzed by IHC at passage 1 compared to respective input biopsies, regardless of implantation variables and clinical features of patients. The Ki-67 proliferation marker colocalized with LANA more frequently in PDXs. Spatial transcriptome analysis revealed increased expression of viral transcripts from latent and lytic gene classes in the PDX. The expanded KSHV+ regions of the PDX maintained signature gene expression of KS tumors, with enrichment in pathways associated with angiogenesis and endothelium development. Cells with characteristics of tumor-associated fibroblasts derived from PDX were propagated for 15 passages. These fibroblast-like cells were permissive for de novo KSHV infection, and one lineage produced CXCL12, a cancer-promoting chemokine. Spatial analysis revealed that fibroblasts are a likely source of CXCL12 signaling to CXCR4 that was upregulated in KS regions. The reproducible expansion of KSHV-infected endothelial cells in PDX from multiple donors and recapitulation of a KS tumor gene signature supports the application of patient-derived KS mouse models for studies of pathogenesis and novel therapies.
Collapse
Affiliation(s)
- Xiaofan Li
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Zoë Weaver Ohler
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Amanda Day
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Laura Bassel
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Anna Grosskopf
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Bahman Afsari
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Takanobu Tagawa
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Wendi Custer
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Ralph Mangusan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Kathryn Lurain
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Joseph Ziegelbauer
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Ramya Ramaswami
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Laurie T. Krug
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| |
Collapse
|
2
|
Mohanty S, Kumar A, Das P, Sahu SK, Mukherjee R, Ramachandranpillai R, Nair SS, Choudhuri T. Nm23-H1 induces apoptosis in primary effusion lymphoma cells via inhibition of NF-κB signaling through interaction with oncogenic latent protein vFLIP K13 of Kaposi’s sarcoma-associated herpes virus. Cell Oncol (Dordr) 2022; 45:967-989. [DOI: 10.1007/s13402-022-00701-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2022] [Indexed: 11/03/2022] Open
|
3
|
Bojagora A, Saridakis V. USP7 manipulation by viral proteins. Virus Res 2020; 286:198076. [DOI: 10.1016/j.virusres.2020.198076] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/14/2020] [Accepted: 06/24/2020] [Indexed: 01/27/2023]
|
4
|
MHC class II fine tuning by ubiquitination: lesson from MARCHs. Immunogenetics 2018; 71:197-201. [DOI: 10.1007/s00251-018-1094-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022]
|
5
|
Watanabe T, Sugimoto A, Hosokawa K, Fujimuro M. Signal Transduction Pathways Associated with KSHV-Related Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1045:321-355. [PMID: 29896674 DOI: 10.1007/978-981-10-7230-7_15] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Signal transduction pathways play a key role in the regulation of cell growth, cell differentiation, cell survival, apoptosis, and immune responses. Bacterial and viral pathogens utilize the cell signal pathways by encoding their own proteins or noncoding RNAs to serve their survival and replication in infected cells. Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is classified as a rhadinovirus in the γ-herpesvirus subfamily and was the eighth human herpesvirus to be discovered from Kaposi's sarcoma specimens. KSHV is closely associated with an endothelial cell malignancy, Kaposi's sarcoma, and B-cell malignancies, primary effusion lymphoma, and multicentric Castleman's disease. Recent studies have revealed that KSHV manipulates the cellular signaling pathways to achieve persistent infection, viral replication, cell proliferation, anti-apoptosis, and evasion of immune surveillance in infected cells. This chapter summarizes recent developments in our understanding of the molecular mechanisms used by KSHV to interact with the cell signaling machinery.
Collapse
Affiliation(s)
- Tadashi Watanabe
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Atsuko Sugimoto
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kohei Hosokawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan.
| |
Collapse
|
6
|
Abstract
Zoonosis is the leading cause of emerging infectious diseases. In a recent article, R. S. Shabman et al. (mSphere 1[1]:e00070-15, 2016, http://dx.doi.org/10.1128/mSphere.00070-15) report the identification of a novel gammaherpesvirus in a cell line derived from the microbat Myotis velifer incautus. This is the first report on a replicating, infectious gammaherpesvirus from bats. The new virus is named bat gammaherpesvirus 8 (BGHV8), also known as Myotis gammaherpesvirus 8, and is able to infect multiple cell lines, including those of human origin. Using next-generation sequencing technology, the authors constructed a full-length annotated genomic map of BGHV8. Phylogenetic analysis of several genes from BGHV8 revealed similarity to several mammalian gammaherpesviruses, including Kaposi’s sarcoma-associated herpesvirus (KSHV).
Collapse
|
7
|
Abstract
Molecular piracy is a biological phenomenon in which one replicon (the pirate) uses the structural proteins encoded by another replicon (the helper) to package its own genome and thus allow its propagation and spread. Such piracy is dependent on a complex web of interactions between the helper and the pirate that occur at several levels, from transcriptional control to macromolecular assembly. The best characterized examples of molecular piracy are from the E. coli P2/P4 system and the S. aureus SaPI pathogenicity island/helper system. In both of these cases, the pirate element is mobilized and packaged into phage-like transducing particles assembled from proteins supplied by a helper phage that belongs to the Caudovirales order of viruses (tailed, dsDNA bacteriophages). In this review we will summarize and compare the processes that are involved in molecular piracy in these two systems.
Collapse
Affiliation(s)
- Gail E. Christie
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, PO Box 980678, Richmond, VA 23298-0678, USA
| | - Terje Dokland
- Department of Microbiology, University of Alabama at Birmingham, 845 19th St South BBRB 311, Birmingham, AL 35294 USA
| |
Collapse
|
8
|
Ray A, Marshall V, Uldrick T, Leighty R, Labo N, Wyvill K, Aleman K, Polizzotto MN, Little RF, Yarchoan R, Whitby D. Sequence analysis of Kaposi sarcoma-associated herpesvirus (KSHV) microRNAs in patients with multicentric Castleman disease and KSHV-associated inflammatory cytokine syndrome. J Infect Dis 2012; 205:1665-76. [PMID: 22448005 PMCID: PMC3415855 DOI: 10.1093/infdis/jis249] [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: 09/27/2011] [Accepted: 12/05/2011] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Kaposi sarcoma-associated herpesvirus (KSHV) encodes 12 pre-microRNAs that yield 25 mature microRNAs. We previously reported phylogenetic analysis of the microRNA-coding region of KSHV from Kaposi sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman disease (MCD) patients. We observed a high level of conservation for most sequences but also a divergent cluster of 5 KSHV sequences, including 2 from MCD patients. METHODS KSHV microRNA sequences from 23 MCD patients and 7 patients with a newly described KSHV-associated inflammatory cytokine syndrome (KICS) were examined by amplification, cloning, and sequencing of a 646-bp fragment of K12/T0.7 encoding microRNA-K12-10 and microRNA-K12-12 and a 2.8-kbp fragment containing the remaining 10 pre-microRNAs. RESULTS Phylogenetic analysis showed a distinct variant cluster consisting exclusively of MCD and KICS patients in all trees. Pearson χ(2) analysis revealed that 40 single-nucleotide polymorphisms (SNPs) at various loci were significantly associated with MCD and KICS risk. Cluster analysis of these SNPs generated several combinations of 3 SNPs as putative indicators of MCD and KICS risk. CONCLUSIONS These findings show that MCD and KICS patients frequently have unusual KSHV microRNA sequences and suggest an association between the observed sequence variation and risk of MCD and KICS.
Collapse
Affiliation(s)
- Alex Ray
- Viral Oncology Section, AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute (NCI)-Frederick
| | - Vickie Marshall
- Viral Oncology Section, AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute (NCI)-Frederick
| | | | | | - Nazzarena Labo
- Viral Oncology Section, AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute (NCI)-Frederick
| | | | | | | | | | | | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute (NCI)-Frederick
| |
Collapse
|
9
|
Cooperation between viral interferon regulatory factor 4 and RTA to activate a subset of Kaposi's sarcoma-associated herpesvirus lytic promoters. J Virol 2011; 86:1021-33. [PMID: 22090118 DOI: 10.1128/jvi.00694-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The four Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded interferon (IFN) regulatory factor homologues (vIRF1 to vIRF4) are used to counter innate immune defenses and suppress p53. The vIRF genes are arranged in tandem but differ in function and expression. In KSHV-infected effusion lymphoma lines, K10.5/vIRF3 and K11/vIRF2 mRNAs are readily detected during latency, whereas K9/vIRF1 and K10/vIRF4 mRNAs are upregulated during reactivation. Here we show that the K10/vIRF4 promoter responds to the lytic switch protein RTA in KSHV-infected cells but is essentially unresponsive in uninfected cells. Coexpression of RTA with vIRF4 is sufficient to restore regulation, a property not shared by other vIRFs. The K9/vIRF1 promoter behaves similarly, and production of infectious virus is enhanced by the presence of vIRF4. Synergy requires the DNA-binding domain (DBD) and C-terminal IRF homology regions of vIRF4. Mutations of arginine residues within the putative DNA recognition helix of vIRF4 or the invariant cysteines of the adjacent CxxC motif abolish cooperation with RTA, in the latter case by preventing self-association. The oligomerization and transactivation functions of RTA are also essential for synergy. The K10/vIRF4 promoter contains two transcription start sites (TSSs), and a 105-bp fragment containing the proximal promoter is responsive to vIRF4/RTA. Binding of a cellular factor(s) to this fragment is altered when both viral proteins are present, suggesting a possible mechanism for transcriptional synergy. Reliance on coregulators encoded by either the host or viral genome provides an elegant strategy for expanding the regulatory potential of a master regulator, such as RTA.
Collapse
|
10
|
Lin LT, Dawson PWH, Richardson CD. Viral interactions with macroautophagy: a double-edged sword. Virology 2010; 402:1-10. [PMID: 20413139 PMCID: PMC7111941 DOI: 10.1016/j.virol.2010.03.026] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 02/18/2010] [Accepted: 03/16/2010] [Indexed: 02/07/2023]
Abstract
Autophagy is a conserved eukaryotic mechanism that mediates the removal of long-lived cytoplasmic macromolecules and damaged organelles via a lysosomal degradative pathway. Recently, a multitude of studies have reported that viral infections may have complex interconnections with the autophagic process. These observations strongly imply that autophagy has virus-specific roles relating to viral replication, host innate and adaptive immune responses, virus-induced cell death programs, and viral pathogenesis. Autophagy can supply internal membrane structures necessary for viral replication or may prolong cell survival during viral infections and postpone cell death. It can influence the survival of both infected and bystander cells. This process has also been linked to the recognition of viral signature molecules during innate immunity and has been suggested to help rid the cell of infection. This review discusses interactions between different viruses and the autophagy pathway, and surveys the current state of knowledge and emerging themes within this field.
Collapse
Affiliation(s)
- Liang-Tzung Lin
- Department of Microbiology and Immunology, Dalhousie University, 5850 College St., 7th Floor Sir Charles Tupper Bldg., Halifax, Nova Scotia, Canada B3H 1X5
| | | | | |
Collapse
|
11
|
Ishido S, Matsuki Y, Goto E, Kajikawa M, Ohmura-Hoshino M. MARCH-I: a new regulator of dendritic cell function. Mol Cells 2010; 29:229-32. [PMID: 20213309 DOI: 10.1007/s10059-010-0051-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 01/20/2010] [Indexed: 01/09/2023] Open
Abstract
We and other groups have demonstrated that the expression level of MHC class II (MHC II) is regulated through ubiquitination of the MHC II beta chain. We also reported that MARCH-I, an E3 ubiquitin ligase, is critical for this process. At present, however, the importance of MARCH-I-mediated MHC II regulation in vivo is still unknown. In this review, we will summarize recent advances in our understanding of MARCH-I-mediated MHC II ubiquitination, and discuss how we can overcome the difficulties inherent in these studies.
Collapse
Affiliation(s)
- Satoshi Ishido
- Laboratory for Infectious Immunity, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, 230-0045, Japan.
| | | | | | | | | |
Collapse
|
12
|
Cai Q, Verma SC, Lu J, Robertson ES. Molecular biology of Kaposi's sarcoma-associated herpesvirus and related oncogenesis. Adv Virus Res 2010; 78:87-142. [PMID: 21040832 PMCID: PMC3142360 DOI: 10.1016/b978-0-12-385032-4.00003-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Kaposi's Sarcoma-associated Herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is the most recently identified human tumor virus,and is associated with the pathogenesis of Kaposi's sarcoma and two lymphoproliferative disorders known to occur frequently in AIDS patients-primary effusion lymphoma and multicentric Castleman disease. In the 15 years since its discovery, intense studies have demonstrated an etiologic role for KSHV in the development of these malignancies. Here, we review the recent advances linked to understanding KSHV latent and lytic life cycle and the molecular mechanisms of KSHV-mediated oncogenesis in terms of transformation, cell signaling, cell growth and survival, angiogenesis, immune invasion and response to microenvironmental stress, and highlight the potential therapeutic targets for blocking KSHV tumorigenesis.
Collapse
Affiliation(s)
- Qiliang Cai
- Department of Microbiology, Abramson, Comprehensive Cancer Center, University of Pennsylvania Medical School, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
13
|
Berkova Z, Wang S, Wise JF, Maeng H, Ji Y, Samaniego F. Mechanism of Fas signaling regulation by human herpesvirus 8 K1 oncoprotein. J Natl Cancer Inst 2009; 101:399-411. [PMID: 19276446 DOI: 10.1093/jnci/djn516] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Herpesvirus 8 (HHV-8) oncoprotein K1 is linked to lymphoproliferation and suppression of apoptosis mediated by the Fas death receptor. Expression of K1 in transgenic mice induces accumulation of lymphoid tissue cells and lymphoma. METHODS To examine how K1 and Fas interact to suppress apoptosis, K1-Fas binding was studied in human embryonic kidney (HEK) and lymphoma (BJAB) cells that expressed wild-type K1 or a K1 Ig domain deletion mutant and were treated with Fas ligand (FasL) or an agonistic Fas antibody, using immunoprecipitation and Western blot analysis. Cleavage of caspase-3 and apoptosis was compared in liver samples from mice that were transfected with empty vector vs with plasmids expressing wild-type K1 or a K1 Ig deletion mutant and treated with agonistic Fas antibody for 7 hours. These studies used immunohistochemical staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay. All statistical tests were two-sided. RESULTS Immunoprecipitation and Western blot analysis of transfected HEK and BJAB cells revealed that wild-type K1 but not Ig-deleted K1 binds to Fas and prevents Fas activation by FasL or by an agonistic Fas antibody. More mice that were transfected with wild-type K1 (7 of 10) than mice transfected with empty vector (3 of 13) or the K1 Ig deletion mutant (0 of 6) survived treatment with the agonistic Fas antibody. Compared with vector-transfected mice, livers of wild-type K1-transfected mice contained fewer cells in which caspase-3 was cleaved (87.6% vs 58.0%, difference = 29.6%, 95% confidence interval [CI] = 19.2% to 40.0%; P = .003) and fewer apoptotic cells (83.7% vs 34.2%, difference = 49.5%, 95% CI = 39.8% to 59.2%; P = .003). CONCLUSIONS K1 blocks Fas signaling by directly binding to Fas through the Ig-like domain of K1 and preventing binding of FasL. The relative resistance of cancer cells to Fas-mediated apoptosis may be due to the inhibition of Fas by Ig domain-containing proteins.
Collapse
Affiliation(s)
- Zuzana Berkova
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, 77054, USA
| | | | | | | | | | | |
Collapse
|
14
|
E3 ubiquitin ligases for MHC molecules. Curr Opin Immunol 2009; 21:78-83. [DOI: 10.1016/j.coi.2009.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Accepted: 01/15/2009] [Indexed: 12/20/2022]
|
15
|
Stebbing J, Powles T, Nelson M, Bower M. Significance of variation within HIV, EBV, and KSHV subtypes. ACTA ACUST UNITED AC 2008; 5:93-102. [PMID: 16928877 DOI: 10.1177/1545109706290171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Since their initial transmission to humans, viruses have diversified extensively through recombination and mutation. The presence of intra- and inter-individual viral diversity influences disease progression, drug resistance, and therapy and presumably explains the conflicting results in many studies, including the failure of peptide-based vaccination strategies. Although HIV is a small RNA virus, coinfection with large DNA viruses, notably the oncogenic gamma-herpesviridae human herpesvirus-8 and Epstein Barr virus, is common. This coinfection occurs secondary to immunosuppression and shared transmission routes with high-risk predisposing behavior. In addition, all 3 of these viruses can lead to chronic infections, long periods of latency, and reactivation characterized by pain and suffering. The efficient targeting of their genetic diversity represents one of the major challenges in their control, both in prophylactic and therapeutic strategies. An understanding of diversity will help delineate whether population-specific vaccine strategies are necessary.
Collapse
Affiliation(s)
- Justin Stebbing
- Department of HIV Medicine, The Chelsea and Westminster Hospital, 369 Fulham Road, London, United Kingdom
| | | | | | | |
Collapse
|
16
|
Stebbing J, Pantanowitz L, Dayyani F, Sullivan RJ, Bower M, Dezube BJ. HIV-associated multicentric Castleman's disease. Am J Hematol 2008; 83:498-503. [PMID: 18260115 DOI: 10.1002/ajh.21137] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Multicentric Castleman's disease (MCD), a relatively rare lymphoproliferative disorder that presents with heterogenous symptoms including fevers, anemia, and multifocal lymphadenopathy, is today most commonly observed in individuals infected with human immunodeficiency virus type-1 (HIV). In such individuals, a lymph node biopsy typically identifies cells that stain for Kaposi's sarcoma-associated herpesvirus proteins, and most HIV-associated MCD features can be attributed to the presence of this gamma-herpesvirus. Surgery and antiviral therapies including highly active antiretroviral therapy, interferon-alpha, foscarnet, ganciclovir, and antibodies to interleukin-6 have proved largely ineffective, and chemotherapy in HIV positive individuals is complicated by limited efficacy and pronounced toxicity. While no randomized trials have been performed, more recently the use of the anti-CD20 monoclonal antibody rituximab in large single center cohorts has been associated with prolonged remissions, radiologic responses, as well as hematologic and serum chemistry normalization of the inflammatory picture observed, at the expense of B cell depletion and flare of Kaposi's sarcoma. MCD represents a model of disease at the interplay between tumor biology, infection, and immunology.
Collapse
Affiliation(s)
- Justin Stebbing
- Department of Medical Oncology, Imperial College School of Science, Technology and Medicine, The Hammersmith Hospitals NHS Trust, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
17
|
Lim CS, Seet BT, Ingham RJ, Gish G, Matskova L, Winberg G, Ernberg I, Pawson T. The K15 protein of Kaposi's sarcoma-associated herpesvirus recruits the endocytic regulator intersectin 2 through a selective SH3 domain interaction. Biochemistry 2007; 46:9874-85. [PMID: 17696407 DOI: 10.1021/bi700357s] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus, also known as human herpesvirus 8, is closely associated with several cancers including Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. The rightmost end of the KSHV genome encodes a protein, K15, with multiple membrane-spanning segments and an intracellular carboxy-terminal tail that contains several conserved motifs with the potential to recruit interaction domains (i.e., SH2, SH3, TRAF) of host cell proteins. K15 has been implicated in downregulating B cell receptor (BCR) signaling through its conserved motifs and may thereby play a role in maintaining viral latency and/or preventing apoptosis of the infected B cells. However, K15's mode of action is largely unknown. We have used mass spectrometry, domain and peptide arrays, and surface plasmon resonance to identify binding partners for a conserved proline-rich sequence (PPLP) in the K15 cytoplasmic tail. We show that the PPLP motif selectively binds the SH3-C domain of an endocytic adaptor protein, Intersectin 2 (ITSN2). This interaction can be observed both in vitro and in cells, where K15 and ITSN2 colocalize to discrete compartments within the B cell. The ability of K15 to associate with ITSN2 suggests a new role for the K15 viral protein in intracellular protein trafficking.
Collapse
Affiliation(s)
- Caesar S Lim
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Pyakurel P, Pak F, Mwakigonja AR, Kaaya E, Biberfeld P. KSHV/HHV-8 and HIV infection in Kaposi's sarcoma development. Infect Agent Cancer 2007; 2:4. [PMID: 17270056 PMCID: PMC1800836 DOI: 10.1186/1750-9378-2-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2006] [Accepted: 02/02/2007] [Indexed: 12/24/2022] Open
Abstract
Kaposi's sarcoma (KS) is a highly and abnormally vascularized tumor-like lesion affecting the skin, lymphnodes and viscera, which develops from early inflammatory stages of patch/plaque to late, nodular tumors composed predominant of spindle cells (SC). These SC are infected with the Kaposi's sarcoma-associated herpesvirus or human herpesvirus-8 (KSHV/HHV-8). KS is promoted during HIV infection by various angiogenic and pro-inflammatory factors including HIV-Tat. The latency associated nuclear antigen type 1 (LANA-1) protein is well expressed in SC, highly immunogenic and considered important in the generation and maintenance of HHV-8 associated malignancies. Various studies favour an endothelial origin of the KS SC, expressing "mixed" lymphatic and vascular endothelial cell markers, possibly representing hybrid phenotypes of endothelial cells (EC). A significant number of SC during KS development are apparently not HHV8 infected, which heterogeneity in viral permissiveness may indicate that non-infected SC may continuously be recruited in to the lesion from progenitor cells and locally triggered to develop permissiveness to HHV8 infection. In the present study various aspects of KS pathogenesis are discussed, focusing on the histopathological as well as cytogenetic and molecular genetic changes in KS.
Collapse
Affiliation(s)
- Pawan Pyakurel
- Immunopathology Lab., Department of Pathology and Oncology, Karolinska Institutet, 171-76 Solna, Stockholm, Sweden
| | - Fatemeh Pak
- Immunopathology Lab., Department of Pathology and Oncology, Karolinska Institutet, 171-76 Solna, Stockholm, Sweden
| | - Amos R Mwakigonja
- Immunopathology Lab., Department of Pathology and Oncology, Karolinska Institutet, 171-76 Solna, Stockholm, Sweden
- Muhimbili University College of Health Sciences, P. O. Box 65023, Dar-Es-Salaam, Tanzania
| | - Ephata Kaaya
- Immunopathology Lab., Department of Pathology and Oncology, Karolinska Institutet, 171-76 Solna, Stockholm, Sweden
- Muhimbili University College of Health Sciences, P. O. Box 65023, Dar-Es-Salaam, Tanzania
| | - Peter Biberfeld
- Immunopathology Lab., Department of Pathology and Oncology, Karolinska Institutet, 171-76 Solna, Stockholm, Sweden
| |
Collapse
|
19
|
Greene W, Kuhne K, Ye F, Chen J, Zhou F, Lei X, Gao SJ. Molecular biology of KSHV in relation to AIDS-associated oncogenesis. Cancer Treat Res 2007; 133:69-127. [PMID: 17672038 PMCID: PMC2798888 DOI: 10.1007/978-0-387-46816-7_3] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
KSHV has been established as the causative agent of KS, PEL, and MCD, malignancies occurring more frequently in AIDS patients. The aggressive nature of KSHV in the context of HIV infection suggests that interactions between the two viruses enhance pathogenesis. KSHV latent infection and lytic reactivation are characterized by distinct gene expression profiles, and both latency and lytic reactivation seem to be required for malignant progression. As a sophisticated oncogenic virus, KSHV has evolved to possess a formidable repertoire of potent mechanisms that enable it to target and manipulate host cell pathways, leading to increased cell proliferation, increased cell survival, dysregulated angiogenesis, evasion of immunity, and malignant progression in the immunocompromised host. Worldwide, approximately 40.3 million people are currently living with HIV infection. Of these, a significant number are coinfected with KSHV. The complex interplay between the two viruses dramatically elevates the risk for development of KSHV-induced malignancies, KS, PEL, and MCD. Although HAART significantly reduces HIV viral load, the entire T-cell repertoire and immune function may not be completely restored. In fact, clinically significant immune deficiency is not necessary for the induction of KSHV-related malignancy. Because of variables such as lack of access to therapy noncompliance with prescribed treatment, failure to respond to treatment and the development of drug-resistant strains of HIV, KSHV-induced malignancies will continue to present as major health concerns.
Collapse
Affiliation(s)
- Whitney Greene
- Tiumor Virology Program, Children's Cancer Research Institute, Department of Pediatrics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Fujimuro M, Hayward SD, Yokosawa H. Molecular piracy: manipulation of the ubiquitin system by Kaposi's sarcoma-associated herpesvirus. Rev Med Virol 2007; 17:405-22. [PMID: 17688306 DOI: 10.1002/rmv.549] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Ubiquitination, one of several post-translational protein modifications, plays a key role in the regulation of cellular events, including protein degradation, signal transduction, endocytosis, protein trafficking, apoptosis and immune responses. Ubiquitin attachment at the lysine residue of cellular factors acts as a signal for endocytosis and rapid degradation by the 26S proteasome. It has recently been observed that viruses, especially oncogenic herpesviruses, utilise molecular piracy by encoding their own proteins to interfere with regulation of cell signalling. Kaposi's sarcoma- associated herpesvirus (KSHV) manipulates the ubiquitin system to facilitate cell proliferation, anti-apoptosis and evasion from immunity. In this review, we will describe the strategies used by KSHV at distinct stages of the viral life-cycle to control the ubiquitin system and promote oncogenesis and viral persistence.
Collapse
Affiliation(s)
- Masahiro Fujimuro
- Faculty of Pharmaceutical Sciences, Department of Biochemistry, Hokkaido University, Kita-ku, Sapporo, Japan.
| | | | | |
Collapse
|
21
|
Ohmura-Hoshino M, Goto E, Matsuki Y, Aoki M, Mito M, Uematsu M, Hotta H, Ishido S. A novel family of membrane-bound E3 ubiquitin ligases. J Biochem 2006; 140:147-54. [PMID: 16954532 DOI: 10.1093/jb/mvj160] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A novel E3 ubiquitin ligase family that consists of viral E3 ubiquitin ligases (E3s) and their mammalian homologues was recently discovered. These novel E3s are membrane-bound molecules that share the secondary structure and catalytic domain for E3 activity. All family members have two transmembrane regions at the center and a RING-CH domain at the amino terminus. Forced expression of these novel E3s has been shown to reduce the surface expression of various membrane proteins through ubiquitination of target molecules. Initial examples of viral E3s were identified in Kaposi's sarcoma associated herpesvirus (KSHV) and murine gamma-herpesvirus 68 (MHV-68) and have been designated as modulator of immune recognition (MIR) 1, 2 and mK3, respectively. MIR 1, 2 and mK3 are able to down-regulate MHC class I molecule expression, and mK3 is required to establish an effective latent viral infection in vivo. The first characterized mammalian homologue to MIR 1, 2 and mK3 is c-MIR/MARCH VIII. Forced expression of c-MIR/MARCH VIII down-regulates B7-2, a co-stimulatory molecule important for antigen presentation. Subsequently, several mammalian molecules related to c-MIR/MARCH VIII have been characterized and named as membrane associated RING-CH (MARCH) family. However, the precise physiological function of MARCH family members remains as yet unknown.
Collapse
Affiliation(s)
- Mari Ohmura-Hoshino
- Laboratory for Infectious Immunity, RIKEN Research Center for Allergy and Immunology 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Since the advent of the HIV-1 pandemic, a close association between HIV-1 infection and the development of selected types of cancers has been brought to light. The discovery of Kaposi sarcoma-associated herpesvirus (KSHV) has led to significant advances in uncovering the virological and molecular mechanisms involved in the pathogenesis of AIDS-related malignancies. Extensive evidence indicates that HIV-1 trans-activating protein Tat plays an oncogenic role in the development of KSHV-associated neoplasms. Comprehensive knowledge of the functions of Tat-1 together with the KSHV genes will contribute to a better understanding of the pathogenesis of virus-associated cancers and the interaction of viruses with their hosts.
Collapse
Affiliation(s)
- Y Aoki
- Development, Astellas Pharma Inc., 17-1 Hasune 3-Chome, Itabashi-ku, 174-8612, Japan.
| | | |
Collapse
|
23
|
Perry ST, Compton T. Kaposi's sarcoma-associated herpesvirus virions inhibit interferon responses induced by envelope glycoprotein gpK8.1. J Virol 2006; 80:11105-14. [PMID: 16956942 PMCID: PMC1642153 DOI: 10.1128/jvi.00846-06] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Kaposi's sarcoma-associated herpesvirus (KSHV) envelope glycoprotein gpK8.1 contributes to cellular attachment through binding cell surface heparan sulfate proteoglycans. By using a soluble recombinant form of gpK8.1, we discovered that a consequence of gpK8.1 interaction with human fibroblasts is the induction of an antiviral response, as characterized by the activation of interferon regulatory factor 3 (IRF-3), production of interferon beta (IFN-beta), and expression of interferon-stimulated antiviral genes. In contrast, neither IFN-beta expression nor a functional antiviral response is observed in cells treated with KSHV virions. The interferon response induced by soluble gpK8.1 can be inhibited by simultaneous treatment with UV-inactivated virions, while the induction of an indicator inflammatory cytokine, interleukin-6, was readily evident in the response to both gpK8.1 and KSHV. In addition, KSHV virions abrogate gpK8.1-mediated activation of IRF-3, an early transcriptional regulator for cellular antiviral responses. Although innate immune responses are initiated during contact between gpK8.1 and cellular receptor(s), these results suggest that the virion contains one or more structural elements that selectively repress an effective antiviral response while allowing cellular responses favorable to the KSHV life cycle.
Collapse
Affiliation(s)
- Stuart T Perry
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, 1400 University Avenue, Madison, WI 53706-1599, USA
| | | |
Collapse
|
24
|
Seo T, Park J, Choe J. Kaposi's Sarcoma–Associated Herpesvirus Viral IFN Regulatory Factor 1 Inhibits Transforming Growth Factor-β Signaling. Cancer Res 2005; 65:1738-47. [PMID: 15753369 DOI: 10.1158/0008-5472.can-04-2374] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus, also called human herpesvirus 8, has been implicated in the pathogenesis of Kaposi's sarcoma, body cavity-based primary effusion lymphoma, and some forms of multicentric Castleman's disease. The Kaposi's sarcoma-associated herpesvirus open reading frame K9 encodes viral IFN regulatory factor 1 (vIRF1), which functions as a repressor of IFN-mediated signal transduction. vIRF1 expression in NIH 3T3 cells leads to transformation and consequently induces malignant fibrosarcoma in nude mice, suggesting that vIRF1 is a strong oncoprotein. Here, we show that vIRF1 inhibited transforming growth factor-beta (TGF-beta) signaling via its targeting of Smad proteins. vIRF1 suppressed TGF-beta-mediated transcription and growth arrest. vIRF1 directly interacted with both Smad3 and Smad4, resulting in inhibition of their transactivation activity. Studies using vIRF1 deletion mutants showed that the central region of vIRF1 was required for vIRF1 association with Smad3 and Smad4 and that this region was also important for inhibition of TGF-beta signaling. In addition, we found that vIRF1 interfered with Smad3-Smad4 complex formation and inhibited Smad3/Smad4 complexes from binding to DNA. These results indicate that vIRF1 inhibits TGF-beta signaling via interaction with Smads. In addition, the data indicate the TGF-beta pathway is an important target for viral oncoproteins.
Collapse
Affiliation(s)
- Taegun Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | | | | |
Collapse
|
25
|
Montaner S, Sodhi A, Servitja JM, Ramsdell AK, Barac A, Sawai ET, Gutkind JS. The small GTPase Rac1 links the Kaposi sarcoma–associated herpesvirus vGPCR to cytokine secretion and paracrine neoplasia. Blood 2004; 104:2903-11. [PMID: 15231571 DOI: 10.1182/blood-2003-12-4436] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractKaposi sarcoma (KS) is a multifocal angioproliferative neoplasm strictly dependent on angiogenic growth factors and cytokines and invariably associated with infection by the Kaposi sarcoma–associated herpesvirus (KSHV or HHV8). A G protein–coupled receptor encoded by KSHV (vGPCR) is able to initiate KS-like tumors when targeted to the vascular endothelium of mice. Analogous to human KS, vGPCR sarcomagenesis involves the paracrine secretion of angiogenic growth factors and proinflammatory molecules from vGPCR-expressing cells. Here we demonstrate that vGPCR up-regulates expression and secretion of critical KS cytokines by stimulating key transcription factors, including nuclear factor–κB (NF-κB), activator protein-1 (AP-1), and nuclear factor of activated T cells (NFAT), through the activation of the small G protein Rac1. Inhibition of Rac1 blocked vGPCR-induced transcription and secretion of KS cytokines, including interleukin-6 (IL-6), IL-8, and growth-regulated oncogene α (GROα), in vitro and reduced vGPCR tumorigenesis in vivo. Moreover, endothelial-specific infection with the constitutively active Rac1QL induced vascular lesions in mice that were remarkably similar to early vGPCR experimental lesions. These results identify Rac1 as a key mediator of vGPCR paracrine neoplasia, suggesting that this small G protein and its downstream effectors may represent suitable therapeutic targets for the treatment of KS.
Collapse
MESH Headings
- Animals
- Cell Line
- Cytokines/biosynthesis
- Cytokines/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/virology
- Herpesvirus 8, Human/chemistry
- Herpesvirus 8, Human/physiology
- Mice
- Mice, Transgenic
- Mutation, Missense
- Paracrine Communication
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/physiology
- Sarcoma, Kaposi/etiology
- Sarcoma, Kaposi/pathology
- Sarcoma, Kaposi/virology
- Transcription Factors/metabolism
- Transcription, Genetic
- Up-Regulation/genetics
- Viral Proteins/genetics
- Viral Proteins/physiology
- rac1 GTP-Binding Protein/metabolism
- rac1 GTP-Binding Protein/physiology
Collapse
Affiliation(s)
- Silvia Montaner
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Dr, Bldg 30, Rm 212, Bethesda, MD 20892-4330, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Fujimuro M, Hayward SD. Manipulation of glycogen-synthase kinase-3 activity in KSHV-associated cancers. J Mol Med (Berl) 2004; 82:223-31. [PMID: 14991150 DOI: 10.1007/s00109-003-0519-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Accepted: 11/26/2003] [Indexed: 12/12/2022]
Abstract
The Kaposi's sarcoma-associated herpesvirus, KSHV, is associated with cancers that have increased incidence in patients who are also HIV positive or who have undergone organ transplantation. It has recently been observed that beta-catenin is overexpressed in two KSHV-associated cancers, Kaposi's sarcoma and primary effusion lymphoma. Investigation of the underlying defect in beta-catenin regulation revealed that the KSHV-encoded LANA protein stabilizes beta-catenin by binding to the negative regulator GSK-3, causing a cell-cycle-dependent nuclear accumulation of GSK-3. Thus, redistribution of GSK-3 has been identified as yet another mechanism through which beta-catenin can be dysregulated and contribute to human cancer.
Collapse
Affiliation(s)
- Masahiro Fujimuro
- Viral Oncology Program, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans St, Baltimore, MD 21231, USA
| | | |
Collapse
|
27
|
Goto E, Ishido S, Sato Y, Ohgimoto S, Ohgimoto K, Nagano-Fujii M, Hotta H. c-MIR, a human E3 ubiquitin ligase, is a functional homolog of herpesvirus proteins MIR1 and MIR2 and has similar activity. J Biol Chem 2003; 278:14657-68. [PMID: 12582153 DOI: 10.1074/jbc.m211285200] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Kaposi's sarcoma associated-herpes virus encodes two proteins, MIR (modulator of immune recognition) 1 and 2, which are involved in the evasion of host immunity. MIR1 and 2 have been shown to function as an E3 ubiquitin ligase for immune recognition-related molecules (e.g. major histocompatibility complex class I, B7-2, and ICAM-1) through the BKS (bovine herpesvirus 4, Kaposi's sarcoma associated-herpes virus, and Swinepox virus) subclass of plant homeodomain (PHD) domain, termed the BKS-PHD domain. Here we show that the human genome also encodes a novel BKS-PHD domain-containing protein that functions as an E3 ubiquitin ligase and whose putative substrate is the B7-2 co-stimulatory molecule. This novel E3 ubiquitin ligase was designated as c-MIR (cellular MIR) based on its functional and structural similarity to MIR1 and 2. Forced expression of c-MIR induced specific down-regulation of B7-2 surface expression through ubiquitination, rapid endocytosis, and lysosomal degradation of the target molecule. This specific targeting was dependent upon the binding of c-MIR to B7-2. Replacing the BKS-PHD domain of MIR1 with the corresponding domain of c-MIR did not alter MIR1 function. The discovery of c-MIR, a novel E3 ubiquitin ligase, highlights the possibility that viral immune regulatory proteins originated in the host genome and presents unique functions of BKS-PHD domain-containing proteins in mammals.
Collapse
Affiliation(s)
- Eiji Goto
- Division of Microbiology, Department of Genome Sciences, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Bénichou S, Benmerah A. [The HIV nef and the Kaposi-sarcoma-associated virus K3/K5 proteins: "parasites"of the endocytosis pathway]. Med Sci (Paris) 2003; 19:100-6. [PMID: 12836198 DOI: 10.1051/medsci/2003191100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The modulation of plasma membrane proteins involved in the communication with the immune system is a general mechanism developed by viruses to escape the immune response. Most of the studied examples have focused on viral proteins that missort cellular proteins during their biosynthesis. However, an increasing number of examples show that the down-modulation can also be achieved after membrane delivery by targeting into the endocytic pathway. For both human immunodeficiency virus (HIV) and Kaposi sarcoma-associated herpesvirus (KSHV), the proteins required for this process are identified, Nef and K3/K5 respectively. The extensive studies in this field have shown that the mechanisms by which these proteins "parasite" the endocytic pathway are completely different. Nef directly interacts with components of the cellular machinery involved in the vesicular transport between the endocytic compartments, mainly the clathrin adaptor complexes (AP), inducing the misrouting of numerous cellular proteins, including CD4, MHC-I, LIGHT, DC-SIGN, CD28 and MHC-II to the endosomal degradation compartment or the trans Golgi-network. The K3 and K5 proteins from KSHV act by inducing the ubiquitylation of the target proteins, such as CMH-I and B7.2, triggering their internalization and subsequent degradation by the highly conserved Tsg101/vps23 ubiquitin-dependent endosomal pathway. While these findings show that the strategies used by viruses to target cellular proteins to the endocytic pathway are extremely diverse, additional investigations are needed for the complete understanding of the specific roles of Nef and K3/K5 in the physiopathology of HIV and KSHV infections, respectively. In addition, these viral factors represent valuable tools to study the pathway they are perturbing.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- Antigens, Surface/metabolism
- Cysteine Endopeptidases/metabolism
- Endocytosis
- Gammaherpesvirinae/genetics
- Gammaherpesvirinae/physiology
- Gene Products, nef/chemistry
- Gene Products, nef/physiology
- Genes, nef
- HIV Infections/immunology
- HIV Infections/virology
- HIV-1/genetics
- HIV-1/physiology
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/physiology
- Humans
- Immediate-Early Proteins/chemistry
- Immediate-Early Proteins/physiology
- Macromolecular Substances
- Mice
- Multienzyme Complexes/metabolism
- Proteasome Endopeptidase Complex
- Protein Processing, Post-Translational
- Protein Transport/physiology
- Sarcoma, Kaposi/immunology
- Sarcoma, Kaposi/virology
- Structure-Activity Relationship
- Ubiquitin/metabolism
- Viral Proteins/chemistry
- Viral Proteins/physiology
- nef Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- Serge Bénichou
- Département de Maladies Infectieuses, Inserm U.567, Cnrs UMR 8104, Institut Cochin, Bâtiment Gustave Roussy, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France.
| | | |
Collapse
|
29
|
Hardwick JM, Bellows DS. Viral versus cellular BCL-2 proteins. Cell Death Differ 2003; 10 Suppl 1:S68-76. [PMID: 12655348 DOI: 10.1038/sj.cdd.4401133] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2002] [Revised: 07/15/2002] [Accepted: 07/29/2002] [Indexed: 11/08/2022] Open
Abstract
All gamma herpesviruses and a few other viruses encode at least one homologue of the mammalian cell death inhibitor BCL-2. Gamma herpesviruses are associated with human and animal lymphoid and epithelial tumours. However, the role of these viral BCL-2 homologues in the virus replication cycle or in human disease is not known, though recent developments show progress in this area. The structure of viral BCL-2 family protein, KSBcl-2, is similar to that of cellular family members, but viral BCL-2 proteins differ functionally from the cellular proteins, apparently escaping the regulatory mechanisms to which their cellular counterparts are subjected. Thus, exploring the biochemical and biological functions of the viral BCL-2 family proteins will increase our understanding of their role in virus infections and will undoubtedly teach us something about their cellular kin.
Collapse
Affiliation(s)
- J M Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
30
|
O'Farrell C, Vaghefi N, Cantonnet M, Buteau B, Boudinot P, Benmansour A. Survey of transcript expression in rainbow trout leukocytes reveals a major contribution of interferon-responsive genes in the early response to a rhabdovirus infection. J Virol 2002; 76:8040-9. [PMID: 12134009 PMCID: PMC155135 DOI: 10.1128/jvi.76.16.8040-8049.2002] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Virus infections induce changes in the expression of host cell genes. A global knowledge of these modifications should help to better understand the virus/host cell interactions. To obtain a more comprehensive view of the rainbow trout response to a viral infection, we used the subtractive suppressive hybridization methodology in the viral hemorrhagic septicemia model of infection. We infected rainbow trout leukocytes with viral hemorrhagic septicemia virus (VHSV), and total RNA from infected and mock-infected cells was compared at 40 h postinfection. Twenty-four virus-induced genes were ultimately retrieved from the subtracted cDNA library, and their differential expression was further confirmed by semiquantitative reverse transcription-PCR and Northern blot analysis. Among these sequences, three were already described as VHSV-induced genes. Eight sequences with known homologs were extended to full-length cDNA using 5' and 3' rapid amplification of cDNA ends, and they were subsequently divided into three functional subsets. Four genes were homologous to mammalian interferon responsive genes, three were similar to chemo-attractant molecules (CXC chemokine, galectin), and two had nucleic acid binding domains. All of the virus-induced genes were also induced by rainbow trout interferon, indicating that the interferon pathway is the predominant component of the anti-VHSV response. They were also expressed in vivo in experimentally infected fish, indicating their biological relevance in natural infection.
Collapse
Affiliation(s)
- Caroline O'Farrell
- Unité de Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, 78352 Jouy-en-Josas Cedex, France
| | | | | | | | | | | |
Collapse
|
31
|
Barnes BJ, Kellum MJ, Field AE, Pitha PM. Multiple regulatory domains of IRF-5 control activation, cellular localization, and induction of chemokines that mediate recruitment of T lymphocytes. Mol Cell Biol 2002; 22:5721-40. [PMID: 12138184 PMCID: PMC133975 DOI: 10.1128/mcb.22.16.5721-5740.2002] [Citation(s) in RCA: 202] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2002] [Revised: 05/01/2002] [Accepted: 05/13/2002] [Indexed: 11/20/2022] Open
Abstract
Transcription factors of the interferon regulatory factor (IRF) family have been identified as critical mediators of early inflammatory gene transcription in infected cells. We recently determined that, besides IRF-3 and IRF-7, IRF-5 serves as a direct transducer of virus-mediated signaling. In contrast to that mediated by the other two IRFs, IRF-5-mediated activation is virus specific. We show that, in addition to Newcastle disease virus (NDV) infection, vesicular stomatitis virus (VSV) and herpes simplex virus type 1 (HSV-1) infection activates IRF-5, leading to the induction of IFNA gene subtypes that are distinct from subtypes induced by NDV. The IRF-5-mediated stimulation of inflammatory genes is not limited to IFNA since in BJAB/IRF-5-expressing cells IRF-5 stimulates transcription of RANTES, macrophage inflammatory protein 1 beta, monocyte chemotactic protein 1, interleukin-8, and I-309 genes in a virus-specific manner. By transient- transfection assay, we identified constitutive-activation (amino acids [aa] 410 to 489) and autoinhibitory (aa 490 to 539) domains in the IRF-5 polypeptide. We identified functional nuclear localization signals (NLS) in the amino and carboxyl termini of IRF-5 and showed that both of these NLS are sufficient for nuclear translocation and retention in infected cells. Furthermore, we demonstrated that serine residues 477 and 480 play critical roles in the response to NDV infection. Mutation of these residues from serine to alanine dramatically decreased phosphorylation and resulted in a substantial loss of IRF-5 transactivation in infected cells. Thus, this study defines the regulatory phosphorylation sites that control the activity of IRF-5 in NDV-infected cells and provides further insight into the structure and function of IRF-5. It also shows that the range of IRF-5 immunoregulatory target genes includes members of the cytokine and chemokine superfamilies.
Collapse
Affiliation(s)
- Betsy J Barnes
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | |
Collapse
|
32
|
Means RE, Ishido S, Alvarez X, Jung JU. Multiple endocytic trafficking pathways of MHC class I molecules induced by a Herpesvirus protein. EMBO J 2002; 21:1638-49. [PMID: 11927548 PMCID: PMC125361 DOI: 10.1093/emboj/21.7.1638] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The K3 protein of a human tumor-inducing herpesvirus, Kaposi's sarcoma-associated herpesvirus (KSHV), down-regulates major histocompatibility complex (MHC) class I surface expression by increasing the rate of endocytosis. In this report, we demonstrate that the internalization of MHC class I by the K3 protein is the result of multiple, consecutive trafficking pathways that accelerate the endocytosis of class I molecules, redirect them to the trans-Golgi network (TGN), and target MHC class I to the lysosomal compartment. Remarkably, these actions of K3 are functionally and genetically separable; the N-terminal zinc finger motif and the central sorting motif are involved in triggering internalization of MHC class I molecules and redirecting them to the TGN. Subsequently, the C-terminal diacidic cluster region of K3 is engaged in targeting MHC class I molecules to the lysosomal compartment. These results demonstrate a novel trafficking mechanism of MHC class I molecules induced by KSHV K3, which ensures viral escape from host immune effector recognition.
Collapse
Affiliation(s)
| | - Satoshi Ishido
- Department of Microbiology and Molecular Genetics, Division of Tumor Virology and
Department of Pathology, New England Regional Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772, USA and Department of Microbiology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan Corresponding author e-mail: R.E.Means and S.Ishido contributed equally to this work
| | - Xavier Alvarez
- Department of Microbiology and Molecular Genetics, Division of Tumor Virology and
Department of Pathology, New England Regional Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772, USA and Department of Microbiology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan Corresponding author e-mail: R.E.Means and S.Ishido contributed equally to this work
| | - Jae U. Jung
- Department of Microbiology and Molecular Genetics, Division of Tumor Virology and
Department of Pathology, New England Regional Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772, USA and Department of Microbiology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan Corresponding author e-mail: R.E.Means and S.Ishido contributed equally to this work
| |
Collapse
|
33
|
Sharp TV, Wang HW, Koumi A, Hollyman D, Endo Y, Ye H, Du MQ, Boshoff C. K15 protein of Kaposi's sarcoma-associated herpesvirus is latently expressed and binds to HAX-1, a protein with antiapoptotic function. J Virol 2002; 76:802-16. [PMID: 11752170 PMCID: PMC136811 DOI: 10.1128/jvi.76.2.802-816.2002] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The Kaposi's sarcoma-associated herpesvirus (KSHV) (or human herpesvirus 8) open reading frame (ORF) K15 encodes a putative integral transmembrane protein in the same genomic location as latent membrane protein 2A of Epstein-Barr virus. Ectopic expression of K15 in cell lines revealed the presence of several different forms ranging in size from full length, approximately 50 kDa, to 17 kDa. Of these different species the 35- and 23-kDa forms were predominant. Mutational analysis of the initiator AUG indicated that translation initiation from this first AUG is required for K15 expression. Computational analysis indicates that the different forms detected may arise due to proteolytic cleavage at internal signal peptide sites. We show that K15 is latently expressed in KSHV-positive primary effusion lymphoma cell lines and in multicentric Castleman's disease. Using a yeast two-hybrid screen we identified HAX-1 (HS1 associated protein X-1) as a binding partner to the C terminus of K15 and show that K15 interacts with cellular HAX-1 in vitro and in vivo. Furthermore, HAX-1 colocalizes with K15 in the endoplasmic reticulum and mitochondria. The function of HAX-1 is unknown, although the similarity of its sequence to those of Nip3 and Bcl-2 infers a role in the regulation of apoptosis. We show here that HAX-1 can form homodimers in vivo and is a potent inhibitor of apoptosis and therefore represents a new apoptosis regulatory protein. The putative functions of K15 with respect to its interaction with HAX-1 are discussed.
Collapse
Affiliation(s)
- Tyson V Sharp
- The CRC Viral Oncology Group, Wolfson Institute for Biomedical Research, University College London, London, United Kingdom WC1E 6BT
| | | | | | | | | | | | | | | |
Collapse
|