1
|
Xu K, Feng H, Zhao R, Huang Y. Targeting Tetraspanins at Cell Interfaces: Functional Modulation and Exosome-Based Drug Delivery for Precise Disease Treatment. ChemMedChem 2025; 20:e202400664. [PMID: 39415492 DOI: 10.1002/cmdc.202400664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/18/2024]
Abstract
Tetraspanins are key players in various physiological and pathological processes, including malignancy, immune response, fertilization, and infectious disease. Affinity ligands targeting the interactions between tetraspanins and partner proteins are promising for modulating downstream signaling pathways, thus emerging as attractive candidates for interfering related biological functions. Due to the involvement in vesicle biogenesis and cargo trafficking, tetraspanins are also regarded as exosome markers, and become molecular targets for drug loading and delivery. Given the rapid development in these areas, this minireview focuses on recent advances in design and engineering of affinity binders toward tetraspanins including CD63, CD81, and CD9. Their mechanism of actions in modulating protein interactions at cell interfaces and treatment of malignant diseases are discussed. Strategies for constructing exosome-based drug delivery platforms are also reviewed, with emphasis on the important roles of tetraspanins and the affinity ligands. Finally, challenges and future development of tetraspanin-targeting therapy and exosomal drug delivery platforms are also discussed.
Collapse
Affiliation(s)
- Kun Xu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huixia Feng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
2
|
Umotoy JC, Kroon PZ, Man S, van Dort KA, Atabey T, Schriek AI, Dekkers G, Herrera-Carrillo E, Geijtenbeek TB, Heukers R, Kootstra NA, van Gils MJ, de Taeye SW. Inhibition of HIV-1 replication by nanobodies targeting tetraspanin CD9. iScience 2024; 27:110958. [PMID: 39391729 PMCID: PMC11465043 DOI: 10.1016/j.isci.2024.110958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/05/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
HIV-1 alters the dynamics and distribution of tetraspanins, a group of proteins integral to membrane organization, to facilitate both entry and egress. Notably, the tetraspanin CD9 is dysregulated during HIV-1 infection, correlating with multifaceted effects on viral replication. Here, we generated llama-derived nanobodies against CD9 to restrict HIV-1 replication. We immunized llamas with recombinant large extracellular loop of CD9 and identified eight clonally distinct nanobodies targeting CD9, each exhibiting a range of affinities and differential binding to cell surface-expressed CD9. Notably, nanobodies T2C001 and T2C002 demonstrated low nanomolar affinities and exhibited differential sensitivities against endogenous and overexpressed CD9 on the cell surface. Although CD9-directed nanobodies did not impede the early stages of HIV-1 life cycle, they effectively inhibited virus-induced syncytia formation and virus replication in T cells and monocyte-derived macrophages. This discovery opens new avenues for host-targeted therapeutic strategies, potentially augmenting existing antiretroviral treatments for HIV-1.
Collapse
Affiliation(s)
- Jeffrey C. Umotoy
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Pascal Z. Kroon
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Shirley Man
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Karel A. van Dort
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Tugba Atabey
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Angela I. Schriek
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Gillian Dekkers
- QVQ Holding BV, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Elena Herrera-Carrillo
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Teunis B.H. Geijtenbeek
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Raimond Heukers
- QVQ Holding BV, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Neeltje A. Kootstra
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Steven W. de Taeye
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Izumida M, Kakoki K, Hayashi H, Matsuyama T, Kubo Y. Rab3a, a small GTP-binding protein, is required for the stabilization of the murine leukaemia virus Gag protein. Small GTPases 2022; 13:162-182. [PMID: 34180342 PMCID: PMC9707528 DOI: 10.1080/21541248.2021.1939631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We recently identified a CD63-interacting protein to understand the role of CD63 in virion production of the human immunodeficiency virus type 1, and we have found that Rab3a forms a complex with CD63. In this study, we analysed the effect of Rab3a on virion production of the murine leukaemia virus (MLV), which is another member of the retrovirus family. We found that Rab3a silencing induced lysosomal degradation of the MLV Gag protein, and recovery of the Rab3a expression restored the level of the Gag protein through a complex formation of MLV Gag and Rab3a, indicating that Rab3a is required for MLV Gag protein expression. In contrast, CD63 silencing decreased the infectivity of released virions but had no effect on virion production, indicating that CD63 facilitates the infectivity of released MLV particles. Although Rab3a induced CD63 degradation in uninfected cells, the complex of MLV Gag and Rab3a suppressed the Rab3a-mediated CD63 degradation in MLV-infected cells. Finally, we found that the MLV Gag protein interacts with Rab3a to stabilize its own protein and CD63 that facilitates the infectivity of released MLV particles. Considering the involvement of Rab3a in lysosome trafficking to the plasma membrane, it may also induce cell surface transport of the MLV Gag protein.
Collapse
Affiliation(s)
- Mai Izumida
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan,Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsura Kakoki
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,Japan Association for Development of Community Medicine, Urology Department, Omura Municipal Hospital, Nagasaki, Japan
| | - Hideki Hayashi
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,Medical University Research Administrator, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Toshifumi Matsuyama
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,Department of Cancer Stem Cell, Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yoshinao Kubo
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan,Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,CONTACT Yoshinao Kubo Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki852-8523, Japan
| |
Collapse
|
4
|
Liao L, Wu Z, Chen W, Zhang H, Li A, Yan Y, Xie Z, Li H, Lin W, Ma J, Zhang X, Xie Q. Anti-CD81 antibody blocks vertical transmission of avian leukosis virus subgroup J. Vet Microbiol 2021; 264:109293. [PMID: 34883334 DOI: 10.1016/j.vetmic.2021.109293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/11/2021] [Accepted: 11/22/2021] [Indexed: 11/18/2022]
Abstract
Control of ALV-J in breed of chicken is still a serious issue that need more attention to be paid. Vertical transmission of ALV-J often give rise to more adverse pathogenicity. However, the way to elimination of ALV-J underlying vertical transmission remains not-well understood. In addition, effective vaccines or drugs have not been developed to prevent and control the transmission of ALV-J so far. CD81, a member of the tetraspanins superfamily, plays important roles in regulating membrane proteins, facilitating cells adhesion or fusion, and also participates in viral infection. The purpose of this study was to investigate whether antibodies against certain tetraspanins affect infection of ALV-J. Here, we showed that anti-CD81 antibody could inhibit viral RNA and protein level. We also found that anti-CD81 antibody interacts with viral protein p27, p32 and gp37. Moreover, treatment with antibody to CD81 can effectively prevent the vertical transmission of ALV-J in animal model. Collectively, current study provides new avenues for the control of ALV-J transmission.
Collapse
Affiliation(s)
- Liqin Liao
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; College of Science and Engineering, Jinan University, Guangzhou, 510632, PR China
| | - Zhiqiang Wu
- Wen's Group Academy, Wen's Foodstuffs Group Co., Ltd., Xinxing, 527400, Guangdong, PR China
| | - Weiguo Chen
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; College of Science and Engineering, Jinan University, Guangzhou, 510632, PR China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China
| | - Huanmin Zhang
- USDA, Agriculture Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI, 48823, USA
| | - Aijun Li
- College of Science and Engineering, Jinan University, Guangzhou, 510632, PR China
| | - Yiming Yan
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China
| | - Zi Xie
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China
| | - Hongxin Li
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China
| | - Wencheng Lin
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, 510642, PR China
| | - Jingyun Ma
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, 510642, PR China
| | - Xinheng Zhang
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China.
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University & Guangdong Provincial Key Lab of Agro Animal Genomics and Molecular Breeding, Guangzhou, 510642, PR China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou, 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, 510642, PR China.
| |
Collapse
|
5
|
Narayanan M, Kulkarni R, Jiang S, Kashanchi F, Prasad A. Cocaine augments neuro-inflammation via modulating extracellular vesicle release in HIV-1 infected immune cells. Retrovirology 2021; 18:26. [PMID: 34530855 PMCID: PMC8444590 DOI: 10.1186/s12977-021-00570-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/19/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Extracellular Vesicles (EV) recently have been implicated in the pathogenesis of HIV-1 syndromes, including neuroinflammation and HIV-1 associated neurological disorder (HAND). Cocaine, an illicit stimulant drug used worldwide is known to exacerbate these HIV-1 associated neurological syndromes. However, the effects of cocaine on EV biogenesis and roles of EVs in enhancing HIV-1 pathogenesis are not yet well defined. RESULTS Here, we investigated the effects of cocaine on EV biogenesis and release in HIV-1 infected immune cells and explored their roles in elicitation of neuroinflammation. We found that cocaine significantly augmented the release of EVs from uninfected and HIV-1 infected T-cells, DCs and macrophages. Further analysis of the molecular components of EVs revealed enhanced expression of adhesion molecules integrin β1 and LFA-1 in those EVs derived from cocaine treated cells. Intriguingly, in EVs derived from HIV-1 infected cells, cocaine treatment significantly increased the levels of viral genes in EVs released from macrophages and DCs, but not in T-cells. Exploring the molecular mechanism to account for this, we found that DCs and macrophages showed enhanced expression of the cocaine receptor Sigma 1-Receptor compared to T-cells. In addition, we found that cocaine significantly altered the integrity of the RNA-induced silencing complex (RISC) in HIV-1 infected macrophages and DCs compared to untreated HIV-1 infected cells. Characterizing further the molecular mechanisms involved in how cocaine increased EV release, we found that cocaine decreased the expression of the interferon-inducible protein BST-2; this resulted in altered trafficking of intracellular virus containing vesicles and EV biogenesis and release. We also observed EVs released from cocaine treated HIV-1 infected macrophages and DCs enhanced HIV-1 trans-infection to T-cells compared to those from untreated and HIV-1 infected cells. These EVs triggered release of proinflammatory cytokines in human brain microvascular endothelial cells (HBMECs) and altered monolayer integrity. CONCLUSIONS Taken together, our results provide a novel mechanism which helps to elucidate the enhanced prevalence of neurological disorders in cocaine using HIV-1 infected individuals and offers insights into developing novel therapeutic strategies against HAND in these hosts.
Collapse
Affiliation(s)
- Manojkumar Narayanan
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Rutuja Kulkarni
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Shuxian Jiang
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Anil Prasad
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
6
|
Fan ZH, Xu Y, Luo W, He XC, Zheng TT, Zhang JJ, Xu XY, Qin QW, Lee XZ. Molecular cloning and characterization of CD63 in common carp infected with koi herpesvirus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 121:104102. [PMID: 33862099 DOI: 10.1016/j.dci.2021.104102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 06/12/2023]
Abstract
CD63 is a member of the four-transmembrane-domain protein superfamily and is the first characterized tetraspanin protein. In the present study, we cloned the common carp (Cyprinus Carpio) CD63 (ccCD63) sequence and found that the ccCD63 ORF contained 711 bp and encoded a protein of 236 amino acids. Homology analysis revealed that the complete ccCD63 sequence had 84.08% amino acid similarity to CD63 of Sinocyclocheilus anshuiensis. Subcellular localization analysis revealed that ccCD63 was localized in the cytoplasm. Quantitative real-time PCR (qRT-PCR) analysis indicated that ccCD63 was expressed in the gill, intestine, liver, spleen, brain and kidney, with higher expression in spleen and brain tissues than in the other examined tissues. After koi herpesvirus (KHV) infection, these tissues exhibited various expression levels of ccCD63. The expression level was the lowest in the liver and highest in the brain; the expression level in the brain was 8.7-fold higher than that in the liver. Furthermore, knockdown of ccCD63 promoted KHV infection. Moreover, ccCD63 was correlated with the regulation of RIG-I/MAVS/TRAF3/TBK1/IRF3 and may be involved in the antiviral response through the RIG-I viral recognition signalling pathway in a TRAF3/TBK1-dependent manner. Taken together, our results suggested that ccCD63 upregulated the interaction of KHV with the host immune system and suppressed the dissemination of KHV.
Collapse
Affiliation(s)
- Z H Fan
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Y Xu
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - W Luo
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - X C He
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - T T Zheng
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - J J Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - X Y Xu
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Q W Qin
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - X Z Lee
- Joint Laboratory of Guangdong Province and Hong Kong Regions on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
7
|
Huang X, Li Y, He X, Chen Y, Wei W, Yang X, Ma K. Gangliosides and CD82 inhibit the motility of colon cancer by downregulating the phosphorylation of EGFR at different tyrosine sites and signaling pathways. Mol Med Rep 2020; 22:3994-4002. [PMID: 33000220 DOI: 10.3892/mmr.2020.11467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/15/2020] [Indexed: 11/06/2022] Open
Abstract
Previous studies have shown that (GM3), a ganglioside, suppresses hepatoma cell motility and migration by inhibiting phosphorylation of EGFR and the activity of the PI3K/AKT signaling pathway. Therefore, the aim of the present study was to investigate whether the combined treatment of CD82 with gangliosides can exert a synergistic inhibitory effect on cell motility and migration. Epidermal growth factor receptor (EGFR) signaling was studied for its role in the mechanism through which CD82 and gangliosides synergistically inhibit the motility and migration of SW620 human colon adenocarcinoma cells. GM3 and/or GM2 treatment, and/or overexpression of CD82 was performed in SW620 cells. High-performance thin layer chromatography, reverse transcription-quantitative PCR, western blotting and flow cytometry assays were used to confirm the content changes of GM2, GM3 and CD82. In addition, the phosphorylation of EGFR, MAPK and Akt were evaluated by western blot analysis. SW620 cell motility was investigated using wound healing analysis and chemotaxis migration assay. The combination of GM3 and GM2 with CD82 was found to markedly suppress EGF-stimulated SW620 cell motility compared with the individual factors or combination of GM2 or GM3 with CD82 by inhibiting the phosphorylation of EGFR. The results suggested that CD82 in combination with either GM2 or GM3 can exert a synergistic inhibitory effect on cell motility and migration; however, the synergistic mechanisms elicited by GM2 or GM3 with CD82 differ.
Collapse
Affiliation(s)
- Xiaohua Huang
- Department of Clinical Biochemistry, College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ying Li
- Department of Clinical Laboratory, The Second Affiliated Hospital, Dalian, Liaoning 116023, P.R. China
| | - Xin He
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yang Chen
- Department of Clinical Biochemistry, College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Wei Wei
- Department of Clinical Biochemistry, College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Keli Ma
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
8
|
Zhao W, Tao XL, Liu N, Lu HZ, Zheng CH, Li XY, Yu M, Li YG. Host protein CD63 promotes viral RNA replication by interacting with human astrovirus non-structural protein nsP1a/4. J Gen Virol 2019; 100:616-628. [DOI: 10.1099/jgv.0.001231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Wei Zhao
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
- 2Biological Anthropology institute, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Xiao Li Tao
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Nian Liu
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
- 2Biological Anthropology institute, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Heng Zhang Lu
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Chun Hong Zheng
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Xiang Yu Li
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Man Yu
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| | - Yong Gang Li
- 1College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou 121200, Liaoning, PR China
| |
Collapse
|
9
|
Dahmane S, Doucet C, Le Gall A, Chamontin C, Dosset P, Murcy F, Fernandez L, Salas D, Rubinstein E, Mougel M, Nollmann M, Milhiet PE. Nanoscale organization of tetraspanins during HIV-1 budding by correlative dSTORM/AFM. NANOSCALE 2019; 11:6036-6044. [PMID: 30869094 DOI: 10.1039/c8nr07269h] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Membrane partition and remodeling play a key role in numerous cell mechanisms, especially in viral replication cycles where viruses subvert the plasma membrane to enter and escape from the host cell. Specifically assembly and release of HIV-1 particles require specific cellular components, which are recruited to the egress site by the viral protein Gag. We previously demonstrated that HIV-1 assembly alters both partitioning and dynamics of the tetraspanins CD9 and CD81, which are key players in many infectious processes, forming enriched areas where the virus buds. In this study we correlated super resolution microscopy mapping of tetraspanins with membrane topography delineated by atomic force microscopy (AFM) in Gag-expressing cells. We revealed that CD9 is specifically trapped within the nascent viral particles, especially at buds tips, suggesting that Gag mediates CD9 and CD81 depletion from the plasma membrane. In addition, we showed that CD9 is organized as small membrane assemblies of few tens of nanometers that can coalesce upon Gag expression.
Collapse
Affiliation(s)
- Selma Dahmane
- Centre de Biochimie Structurale (CBS), INSERM, CNRS, Univ Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Florin L, Lang T. Tetraspanin Assemblies in Virus Infection. Front Immunol 2018; 9:1140. [PMID: 29887866 PMCID: PMC5981178 DOI: 10.3389/fimmu.2018.01140] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/07/2018] [Indexed: 12/23/2022] Open
Abstract
Tetraspanins (Tspans) are a family of four-span transmembrane proteins, known as plasma membrane “master organizers.” They form Tspan-enriched microdomains (TEMs or TERMs) through lateral association with one another and other membrane proteins. If multiple microdomains associate with each other, larger platforms can form. For infection, viruses interact with multiple cell surface components, including receptors, activating proteases, and signaling molecules. It appears that Tspans, such as CD151, CD82, CD81, CD63, CD9, Tspan9, and Tspan7, coordinate these associations by concentrating the interacting partners into Tspan platforms. In addition to mediating viral attachment and entry, these platforms may also be involved in intracellular trafficking of internalized viruses and assist in defining virus assembly and exit sites. In conclusion, Tspans play a role in viral infection at different stages of the virus replication cycle. The present review highlights recently published data on this topic, with a focus on events at the plasma membrane. In light of these findings, we propose a model for how Tspan interactions may organize cofactors for viral infection into distinct molecular platforms.
Collapse
Affiliation(s)
- Luise Florin
- Department of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
11
|
Kubo Y, Izumida M, Yashima Y, Yoshii-Kamiyama H, Tanaka Y, Yasui K, Hayashi H, Matsuyama T. Gamma-interferon-inducible, lysosome/endosome-localized thiolreductase, GILT, has anti-retroviral activity and its expression is counteracted by HIV-1. Oncotarget 2018; 7:71255-71273. [PMID: 27655726 PMCID: PMC5342076 DOI: 10.18632/oncotarget.12104] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 08/26/2016] [Indexed: 11/25/2022] Open
Abstract
The mechanism by which type II interferon (IFN) inhibits virus replications remains to be identified. Murine leukemia virus (MLV) replication was significantly restricted by γ-IFN, but not human immunodeficiency virus type 1 (HIV-1) replication. Because MLV enters host cells via endosomes, we speculated that certain cellular factors among γ-IFN-induced, endosome-localized proteins inhibit MLV replication. We found that γ-IFN-inducible lysosomal thiolreductase (GILT) significantly restricts HIV-1 replication as well as MLV replication by its thiolreductase activity. GILT silencing enhanced replication-defective HIV-1 vector infection and virion production in γ-IFN-treated cells, although γ-IFN did not inhibit HIV-1 replication. This result showed that GILT is required for the anti-viral activity of γ-IFN. Interestingly, GILT protein level was increased by γ-IFN in uninfected cells and env-deleted HIV-1-infected cells, but not in full-length HIV-1-infected cells. γ-IFN-induced transcription from the γ-IFN-activation sequence was attenuated by the HIV-1 Env protein. These results suggested that the γ-IFN cannot restrict HIV-1 replication due to the inhibition of γ-IFN signaling by HIV-1 Env. Finally, we found that 4,4′-dithiodipyridine (4-PDS), which inhibits S-S bond formation at acidic pH, significantly suppresses HIV-1 vector infection and virion production, like GILT. In conclusion, this study showed that GILT functions as a host restriction factor against the retroviruses, and a GILT mimic, 4-PDS, is the leading compound for the development of novel concept of anti-viral agents.
Collapse
Affiliation(s)
- Yoshinao Kubo
- Division of Cytokine Signaling, Graduate School of Medical Sciences, Nagasaki University, Nagasaki, Japan.,Department of AIDS Research, Institute of Tropical Medicine, G-COE, Nagasaki University, Nagasaki, Japan
| | - Mai Izumida
- Division of Cytokine Signaling, Graduate School of Medical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuka Yashima
- Division of Cytokine Signaling, Graduate School of Medical Sciences, Nagasaki University, Nagasaki, Japan
| | - Haruka Yoshii-Kamiyama
- Division of Cytokine Signaling, Graduate School of Medical Sciences, Nagasaki University, Nagasaki, Japan.,Department of AIDS Research, Institute of Tropical Medicine, G-COE, Nagasaki University, Nagasaki, Japan
| | - Yuetsu Tanaka
- Department of Immunology, Graduate School and Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kiyoshi Yasui
- Division of Cytokine Signaling, Graduate School of Medical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hideki Hayashi
- Division of Cytokine Signaling, Graduate School of Medical Sciences, Nagasaki University, Nagasaki, Japan
| | - Toshifumi Matsuyama
- Division of Cytokine Signaling, Graduate School of Medical Sciences, Nagasaki University, Nagasaki, Japan.,Present address: Department of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Quantitative membrane proteomics reveals a role for tetraspanin enriched microdomains during entry of human cytomegalovirus. PLoS One 2017; 12:e0187899. [PMID: 29121670 PMCID: PMC5679760 DOI: 10.1371/journal.pone.0187899] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/27/2017] [Indexed: 12/15/2022] Open
Abstract
Human cytomegalovirus (HCMV) depends on and modulates multiple host cell membrane proteins during each stage of the viral life cycle. To gain a global view of the impact of HCMV-infection on membrane proteins, we analyzed HCMV-induced changes in the abundance of membrane proteins in fibroblasts using stable isotope labeling with amino acids (SILAC), membrane fractionation and protein identification by two-dimensional liquid chromatography and tandem mass spectrometry. This systematic approach revealed that CD81, CD44, CD98, caveolin-1 and catenin delta-1 were down-regulated during infection whereas GRP-78 was up-regulated. Since CD81 downregulation was also observed during infection with UV-inactivated virus we hypothesized that this tetraspanin is part of the viral entry process. Interestingly, additional members of the tetraspanin family, CD9 and CD151, were also downregulated during HCMV-entry. Since tetraspanin-enriched microdomains (TEM) cluster host cell membrane proteins including known CMV receptors such as integrins, we studied whether TEMs are required for viral entry. When TEMs were disrupted with the cholesterol chelator methyl-β-cylcodextrin, viral entry was inhibited and this inhibition correlated with reduced surface levels of CD81, CD9 and CD151, whereas integrin levels remained unchanged. Furthermore, simultaneous siRNA-mediated knockdown of multiple tetraspanins inhibited viral entry whereas individual knockdown had little effect suggesting essential, but redundant roles for individual tetraspanins during entry. Taken together, our data suggest that TEM act as platforms for receptors utilized by HCMV for entry into cells.
Collapse
|
13
|
Single-cell analysis identifies cellular markers of the HIV permissive cell. PLoS Pathog 2017; 13:e1006678. [PMID: 29073251 PMCID: PMC5658171 DOI: 10.1371/journal.ppat.1006678] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/03/2017] [Indexed: 11/19/2022] Open
Abstract
Cellular permissiveness to HIV infection is highly heterogeneous across individuals. Heterogeneity is also found across CD4+ T cells from the same individual, where only a fraction of cells gets infected. To explore the basis of permissiveness, we performed single-cell RNA-seq analysis of non-infected CD4+ T cells from high and low permissive individuals. Transcriptional heterogeneity translated in a continuum of cell states, driven by T-cell receptor-mediated cell activation and was strongly linked to permissiveness. Proteins expressed at the cell surface and displaying the highest correlation with T cell activation were tested as biomarkers of cellular permissiveness to HIV. FACS sorting using antibodies against several biomarkers of permissiveness led to an increase of HIV cellular infection rates. Top candidate biomarkers included CD25, a canonical activation marker. The combination of CD25 high expression with other candidate biomarkers led to the identification of CD298, CD63 and CD317 as the best biomarkers for permissiveness. CD25highCD298highCD63highCD317high cell population showed an enrichment of HIV-infection of up to 28 fold as compared to the unsorted cell population. The purified hyper-permissive cell subpopulation was characterized by a downregulation of interferon-induced genes and several known restriction factors. Single-cell RNA-seq analysis coupled with functional characterization of cell biomarkers provides signatures of the "HIV-permissive cell".
Collapse
|
14
|
Kim J, Jobe O, Peachman KK, Michael NL, Robb ML, Rao M, Rao VB. Quantitative analyses reveal distinct sensitivities of the capture of HIV-1 primary viruses and pseudoviruses to broadly neutralizing antibodies. Virology 2017; 508:188-198. [PMID: 28577855 DOI: 10.1016/j.virol.2017.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 01/13/2023]
Abstract
Development of vaccines capable of eliciting broadly neutralizing antibodies (bNAbs) is a key goal to controlling the global AIDS epidemic. To be effective, bNAbs must block the capture of HIV-1 to prevent viral acquisition and establishment of reservoirs. However, the role of bNAbs, particularly during initial exposure of primary viruses to host cells, has not been fully examined. Using a sensitive, quantitative, and high-throughput qRT-PCR assay, we found that primary viruses were captured by host cells and converted into a trypsin-resistant form in less than five minutes. We discovered, unexpectedly, that bNAbs did not block primary virus capture, although they inhibited the capture of pseudoviruses/IMCs and production of progeny viruses at 48h. Further, viruses escaped bNAb inhibition unless the bNAbs were present in the initial minutes of exposure of virus to host cells. These findings will have important implications for HIV-1 vaccine design and determination of vaccine efficacy.
Collapse
Affiliation(s)
- Jiae Kim
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA; Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring 20910, MD, USA
| | - Ousman Jobe
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA; Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring 20910, MD, USA
| | - Kristina K Peachman
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA; Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring 20910, MD, USA
| | - Nelson L Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring 20910, MD, USA
| | - Merlin L Robb
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Mangala Rao
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring 20910, MD, USA.
| | - Venigalla B Rao
- The Catholic University of America, Department of Biology, 620 Michigan Ave., NE, Washington DC 20064, USA.
| |
Collapse
|
15
|
Effect of prolonged freezing of semen on exosome recovery and biologic activity. Sci Rep 2017; 7:45034. [PMID: 28338013 PMCID: PMC5364471 DOI: 10.1038/srep45034] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 02/17/2017] [Indexed: 01/07/2023] Open
Abstract
Exosomes are important vehicles of intercellular communication that shape host responses to physiologic, tumorigenic, and pathogenic conditions. The composition and function of exosomes are dynamic and depends on the state and condition of the cellular source. In prior work, we found that semen exosomes (SE) from healthy donors who do not use illicit drugs potently inhibit HIV-1. Following semen donation, specimens are either used immediately or frozen for use at a later time. It has been shown that short-term freezing of semen has no effect on SE-mediated HIV-1 inhibition. However, the effect of illicit drugs and prolonged freezing on SE bioactivity is unknown. Here, we show preservation of SE physical properties, (morphology, concentration, intensity/size) irrespective of illicit drug use or duration of semen freezing. Interestingly, illicit drugs and prolonged freezing decreased the levels of SE-bound CD63/CD9 and acetylcholinesterase activity respectively. Furthermore, we show differential effects of illicit drug use and prolonged freezing on SE-mediated HIV-1 inhibition. Our results highlight the importance of the source of SE and condition of semen storage on SE content and function. In-depth evaluation of donor drug-use and duration of semen storage on SE cargo and bioactivity will advance our understanding of SE composition and function.
Collapse
|
16
|
Jobe O, Trinh HV, Kim J, Alsalmi W, Tovanabutra S, Ehrenberg PK, Peachman KK, Gao G, Thomas R, Kim JH, Michael NL, Alving CR, Rao VB, Rao M. Effect of cytokines on Siglec-1 and HIV-1 entry in monocyte-derived macrophages: the importance of HIV-1 envelope V1V2 region. J Leukoc Biol 2016; 99:1089-106. [PMID: 26667473 PMCID: PMC4952014 DOI: 10.1189/jlb.2a0815-361r] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/29/2015] [Accepted: 11/17/2015] [Indexed: 02/02/2023] Open
Abstract
Monocytes and monocyte-derived macrophages express relatively low levels of CD4. Despite this, macrophages can be effectively infected with human immunodeficiency virus type 1. Macrophages have a critical role in human immunodeficiency virus type 1 transmission; however, the mechanism or mechanisms of virus infection are poorly understood. We report that growth factors, such as granulocyte macrophage colony-stimulating factor and macrophage colony-stimulating factor affect the phenotypic profile and permissiveness of macrophages to human immunodeficiency virus type 1. Human immunodeficiency virus type 1 infection of monocyte-derived macrophages derived from granulocyte macrophage and macrophage colony-stimulating factors was predominantly facilitated by the sialic acid-binding immunoglobulin-like lectin-1. The number of sialic acid-binding immunoglobulin-like lectin receptors on macrophage colony-stimulating factor-derived monocyte-derived macrophages was significantly greater than on granulocyte macrophage colony-stimulating factor-derived monocyte-derived macrophages, and correspondingly, human immunodeficiency virus type 1 infection was greater in the macrophage colony-stimulating factor-derived monocyte-derived macrophages. Single-genome analysis and quantitative reverse transcriptase-polymerase chain reaction revealed that the differences in infectivity was not due to differences in viral fitness or in viral variants with differential infectivity but was due to reduced viral entry into the granulocyte macrophage colony-stimulating factor-derived monocyte-derived macrophages. Anti-sialic acid-binding immunoglobulin-like lectin, trimeric glycoprotein 145, and scaffolded V1V2 proteins were bound to sialic acid-binding immunoglobulin-like lectin and significantly reduced human immunodeficiency virus type 1 entry and infection. Furthermore, sialic acid residues present in the V1V2 region of the envelope protein mediated human immunodeficiency virus type 1 interaction with sialic acid-binding immunoglobulin-like lectin and entry into macrophage colony-stimulating factor-derived monocyte-derived macrophages. Removal of sialic acid residues or glycans from scaffolded V1V2 protein decreased human immunodeficiency virus type 1 infectivity. These results highlight the importance of sialic acids on the V1V2 region in binding to sialic acid-binding immunoglobulin-like lectin and suggest that the unusually long surface-exposed sialic acid-binding immunoglobulin-like lectin might aid in the capture and entry of human immunodeficiency virus type 1 into monocyte-derived macrophages.
Collapse
Affiliation(s)
- Ousman Jobe
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland, USA; Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Hung V Trinh
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland, USA; Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jiae Kim
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland, USA; Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Wadad Alsalmi
- Department of Biology, The Catholic University of America, Washington, DC, USA
| | - Sodsai Tovanabutra
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland, USA; Laboratory of Molecular Virology and Pathogenesis, Viral Sequencing Core, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; and
| | - Philip K Ehrenberg
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland, USA; Host Genetics Section, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; and
| | - Kristina K Peachman
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland, USA; Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Guofen Gao
- Department of Biology, The Catholic University of America, Washington, DC, USA
| | - Rasmi Thomas
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland, USA; Host Genetics Section, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; and
| | - Jerome H Kim
- Laboratory of Molecular Virology and Pathogenesis, Viral Sequencing Core, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; and
| | - Nelson L Michael
- Laboratory of Molecular Virology and Pathogenesis, Viral Sequencing Core, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; and
| | - Carl R Alving
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Venigalla B Rao
- Department of Biology, The Catholic University of America, Washington, DC, USA
| | - Mangala Rao
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA;
| |
Collapse
|
17
|
Fu E, Pan L, Xie Y, Mu D, Liu W, Jin F, Bai X. Tetraspanin CD63 is a regulator of HIV-1 replication. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:1184-1198. [PMID: 25973004 PMCID: PMC4396296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/09/2015] [Indexed: 06/04/2023]
Abstract
Macrophages and CD4(+) T-cells are the major reservoirs for HIV-1 infection. CD63 is a tetraspanin transmembrane protein, which has been shown to play an essential role during HIV-1 replication in macrophages. In this study, we further confirm the requirement of CD63 in HIV-1 replication events in primary human CD4(+) T-cells, dendritic cells, and a CD4(+) cell line. Most interestingly, we also show the evidences for the co-localization and internalization of CD63 and HIV-1 major receptor CD4 in primary human macrophages and CD4(+) cell line by confocal microscopy and Co-Immunoprecipitation assay. Analysis revealed that CD63-depleted CD4(+) T-cells, dendritic cells, and a cell line showed significant decrease in HIV-1 production. Further analysis showed that CD63 down regulation reduced production of the early HIV protein Tat, and affected HIV protein Gag by CD63-Gag interaction. In agreement, CD63 silencing also inhibited production of the late protein p24. Furthermore, we revealed that CD63 silencing has no effect on HIV-1 replication with extensive viral challenge (MOI > 0.2). These findings suggest that CD63 plays a dual-role both in early and late HIV-1 life cycle with a range of HIV-1 infection (MOI < 0.2).
Collapse
Affiliation(s)
- Enqing Fu
- Department of Respiratory and Critical Medicine, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Lei Pan
- Department of Respiratory and Critical Medicine, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Yonghong Xie
- Department of Respiratory and Critical Medicine, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Deguang Mu
- Department of Respiratory and Critical Medicine, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Wei Liu
- Department of Respiratory and Critical Medicine, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Faguang Jin
- Department of Respiratory and Critical Medicine, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Xuefan Bai
- Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| |
Collapse
|
18
|
Murungi EK, Kariithi HM, Adunga V, Obonyo M, Christoffels A. Evolution and Structural Analyses of Glossina morsitans (Diptera; Glossinidae) Tetraspanins. INSECTS 2014; 5:885-908. [PMID: 26462947 PMCID: PMC4592607 DOI: 10.3390/insects5040885] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 10/23/2014] [Accepted: 11/02/2014] [Indexed: 12/25/2022]
Abstract
Tetraspanins are important conserved integral membrane proteins expressed in many organisms. Although there is limited knowledge about the full repertoire, evolution and structural characteristics of individual members in various organisms, data obtained so far show that tetraspanins play major roles in membrane biology, visual processing, memory, olfactory signal processing, and mechanosensory antennal inputs. Thus, these proteins are potential targets for control of insect pests. Here, we report that the genome of the tsetse fly, Glossina morsitans (Diptera: Glossinidae) encodes at least seventeen tetraspanins (GmTsps), all containing the signature features found in the tetraspanin superfamily members. Whereas six of the GmTsps have been previously reported, eleven could be classified as novel because their amino acid sequences do not map to characterized tetraspanins in the available protein data bases. We present a model of the GmTsps by using GmTsp42Ed, whose presence and expression has been recently detected by transcriptomics and proteomics analyses of G. morsitans. Phylogenetically, the identified GmTsps segregate into three major clusters. Structurally, the GmTsps are largely similar to vertebrate tetraspanins. In view of the exploitation of tetraspanins by organisms for survival, these proteins could be targeted using specific antibodies, recombinant large extracellular loop (LEL) domains, small-molecule mimetics and siRNAs as potential novel and efficacious putative targets to combat African trypanosomiasis by killing the tsetse fly vector.
Collapse
Affiliation(s)
- Edwin K Murungi
- South African National Bioinformatics Institute (SANBI), University of the Western Cape, Private Bag X79, Bellville, Cape Town 7535, South Africa.
| | - Henry M Kariithi
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization (KALRO), P.O. Box 57811, Kaptagat Rd, Nairobi 00200, Kenya.
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, Wageningen 6708 PB, The Netherlands.
| | - Vincent Adunga
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Egerton 20115, Kenya.
| | - Meshack Obonyo
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Egerton 20115, Kenya.
| | - Alan Christoffels
- South African National Bioinformatics Institute (SANBI), University of the Western Cape, Private Bag X79, Bellville, Cape Town 7535, South Africa.
| |
Collapse
|
19
|
Li G, Endsley MA, Somasunderam A, Gbota SL, Mbaka MI, Murray JL, Ferguson MR. The dual role of tetraspanin CD63 in HIV-1 replication. Virol J 2014; 11:23. [PMID: 24507450 PMCID: PMC3944621 DOI: 10.1186/1743-422x-11-23] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 02/05/2014] [Indexed: 01/24/2023] Open
Abstract
Background Previously, we showed that the tetraspanin membrane protein CD63 mediates both early and post-integration stages of the HIV-1 replication cycle. The temporal roles of CD63 were discerned using monoclonal antibodies and small interfering RNAs (siRNAs) to block CD63 function, and determining which of the sequential steps in HIV-1 replication were disrupted. Inhibition was shown to occur during early infection, suggestive of involvement in virus entry or reverse transcription. In addition, we have shown that treatment with CD63 siRNA post-infection, significantly inhibited virus production in supernatant, suggesting an important role for CD63 in macrophages during HIV-1 replication events occurring after proviral integration, and possibly during egress. Results In this study we used CD63 siRNA to investigate the infectivity of pseudotyped viruses (carrying an NL4-3 Env-negative luciferase backbone) in primary human macrophages. We demonstrated that lab adapted R5- and R5X4-tropic HIV-1 strains are significantly inhibited by CD63 silencing. However, the infectivity of MLV or VSV-pseudotyped strains, which enter though receptor-mediated endocytosis, is unaffected by silencing CD63. These results indicate that CD63 may support Env-mediated entry or fusion events facilitated though CD4 and CCR5. Also, antibody and siRNA-based CD63 inhibition studies indicate a potential role for CD63 following proviral integration. Further, we show that CD63 expression is key for efficient replication in primary CD4+ T cells, complementing our prior studies with primary human macrophages and immortalized cell lines. Conclusions Collectively, these findings indicate that CD63 may support Env-mediated fusion as well as a late (post-integration) step in the HIV-1 replication cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Monique R Ferguson
- Department of Internal Medicine, Division of Infectious Diseases, School of Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0435, USA.
| |
Collapse
|
20
|
Tippett E, Cameron PU, Marsh M, Crowe SM. Characterization of tetraspanins CD9, CD53, CD63, and CD81 in monocytes and macrophages in HIV-1 infection. J Leukoc Biol 2013; 93:913-20. [PMID: 23570947 DOI: 10.1189/jlb.0812391] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Tetraspanins are a family of membrane-organizing proteins that mediate diverse functions. Little is known of their expression or function in myeloid cells. Here, expression of CD9, CD53, CD63, and CD81, tetraspanins that have been implicated in HIV-1 pathogenesis, were characterized in normal monocyte subsets, in MDM, and in HIV-1-infected donors. We show that tetraspanins are expressed differentially by monocyte subsets, with higher CD9 and CD63 and lower CD53 and CD81 levels on CD14++CD16- monocytes compared with CD14++CD16+ and CD14+CD16++ subsets. Maturation of monocytes resulted in increased CD9 expression and apparent relocation of CD63 and CD53 from surface to intracellular membranes. Expression was modulated by cytokines, and CD9 was a marker of anti-inflammatory and CD53 a marker of proinflammatory MDM. Tetraspanin expression on monocyte subsets from HIV-1-infected donors receiving antiretroviral therapy was unchanged compared with that in uninfected donors. However, CD53 expression was inversely correlated with viral load in HIV-1-infected donors not on therapy. This study is the first to comprehensively characterize tetraspanin expression on monocyte subsets and macrophages in health and during HIV-1 infection. It demonstrates regulation of tetraspanin expression by cytokines, and CD53 expression as a novel correlate of a proinflammatory phenotype. This paper characterizes tetraspanins in myeloid cells and shows that tetraspanins are expressed differentially in monocyte subsets and are modified in inflammatory conditions.
Collapse
Affiliation(s)
- Emma Tippett
- Centre for Virology, The Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
21
|
Friedrich BM, Dziuba N, Li G, Endsley MA, Murray JL, Ferguson MR. Host factors mediating HIV-1 replication. Virus Res 2011; 161:101-14. [PMID: 21871504 DOI: 10.1016/j.virusres.2011.08.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 08/05/2011] [Accepted: 08/08/2011] [Indexed: 10/17/2022]
Abstract
Human immunodeficiency virus type 1(HIV-1) infection is the leading cause of death worldwide in adults attributable to infectious diseases. Although the majority of infections are in sub-Saharan Africa and Southeast Asia, HIV-1 is also a major health concern in most countries throughout the globe. While current antiretroviral treatments are generally effective, particularly in combination therapy, limitations exist due to drug resistance occurring among the drug classes. Traditionally, HIV-1 drugs have targeted viral proteins, which are mutable targets. As cellular genes mutate relatively infrequently, host proteins may prove to be more durable targets than viral proteins. HIV-1 replication is dependent upon cellular proteins that perform essential roles during the viral life cycle. Maraviroc is the first FDA-approved antiretroviral drug to target a cellular factor, HIV-1 coreceptor CCR5, and serves to intercept viral-host protein-protein interactions mediating entry. Recent large-scale siRNA and shRNA screens have revealed over 1000 candidate host factors that potentially support HIV-1 replication, and have implicated new pathways in the viral life cycle. These host proteins and cellular pathways may represent important targets for future therapeutic discoveries. This review discusses critical cellular factors that facilitate the successive steps in HIV-1 replication.
Collapse
Affiliation(s)
- Brian M Friedrich
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas 77555-0435, United States.
| | | | | | | | | | | |
Collapse
|
22
|
Friedrich BM, Murray JL, Li G, Sheng J, Hodge TW, Rubin DH, O'Brien WA, Ferguson MR. A functional role for ADAM10 in human immunodeficiency virus type-1 replication. Retrovirology 2011; 8:32. [PMID: 21569301 PMCID: PMC3118345 DOI: 10.1186/1742-4690-8-32] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 05/11/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Gene trap insertional mutagenesis was used as a high-throughput approach to discover cellular genes participating in viral infection by screening libraries of cells selected for survival from lytic infection with a variety of viruses. Cells harboring a disrupted ADAM10 (A Disintegrin and Metalloprotease 10) allele survived reovirus infection, and subsequently ADAM10 was shown by RNA interference to be important for replication of HIV-1. RESULTS Silencing ADAM10 expression with small interfering RNA (siRNA) 48 hours before infection significantly inhibited HIV-1 replication in primary human monocyte-derived macrophages and in CD4⁺ cell lines. In agreement, ADAM10 over-expression significantly increased HIV-1 replication. ADAM10 down-regulation did not inhibit viral reverse transcription, indicating that viral entry and uncoating are also independent of ADAM10 expression. Integration of HIV-1 cDNA was reduced in ADAM10 down-regulated cells; however, concomitant 2-LTR circle formation was not detected, suggesting that HIV-1 does not enter the nucleus. Further, ADAM10 silencing inhibited downstream reporter gene expression and viral protein translation. Interestingly, we found that while the metalloprotease domain of ADAM10 is not required for HIV-1 replication, ADAM15 and γ-secretase (which proteolytically release the extracellular and intracellular domains of ADAM10 from the plasma membrane, respectively) do support productive infection. CONCLUSIONS We propose that ADAM10 facilitates replication at the level of nuclear trafficking. Collectively, our data support a model whereby ADAM10 is cleaved by ADAM15 and γ-secretase and that the ADAM10 intracellular domain directly facilitates HIV-1 nuclear trafficking. Thus, ADAM10 represents a novel cellular target class for development of antiretroviral drugs.
Collapse
Affiliation(s)
- Brian M Friedrich
- Departments of Pathology and Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - James L Murray
- Zirus, Inc., 1384 Buford Business Boulevard, Suite 700, Buford, GA, 30518, USA
| | - Guangyu Li
- Departments of Pathology and Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Jinsong Sheng
- Departments of Medicine and Microbiology & Immunology, Vanderbilt University, 1161 21st Ave South, Nashville, TN, 37232, USA
| | - Thomas W Hodge
- Zirus, Inc., 1384 Buford Business Boulevard, Suite 700, Buford, GA, 30518, USA
| | - Donald H Rubin
- Departments of Medicine and Microbiology & Immunology, Vanderbilt University, 1161 21st Ave South, Nashville, TN, 37232, USA
- Research Medicine, VA Tennessee Valley Healthcare System, 1310 24th Ave South, Nashville, TN 37212, USA
| | - William A O'Brien
- Departments of Pathology and Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
- Zirus, Inc., 1384 Buford Business Boulevard, Suite 700, Buford, GA, 30518, USA
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, 201 Dowman Dr., Atlanta, GA, 30322, USA
| | - Monique R Ferguson
- Departments of Pathology and Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| |
Collapse
|
23
|
Li G, Dziuba N, Friedrich B, Murray JL, Ferguson MR. A post-entry role for CD63 in early HIV-1 replication. Virology 2011; 412:315-24. [PMID: 21315401 DOI: 10.1016/j.virol.2011.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 12/19/2010] [Accepted: 01/14/2011] [Indexed: 10/18/2022]
Abstract
Macrophages and CD4(+) lymphocytes are the major reservoirs for HIV-1 infection. CD63 is a tetraspanin transmembrane protein, which has been shown to play an essential role during HIV-1 replication in macrophages. In this study, we further confirm the requirement of CD63 in early HIV-1 replication events in both macrophages and a CD4(+) cell line. Further analysis revealed that viral attachment and cell-cell fusion were unaffected by CD63 silencing. However, CD63-depleted macrophages showed a significant decrease in the initiation and completion of HIV-1 reverse transcription, affecting subsequent events of the HIV-1 life cycle. Integration of HIV-1 cDNA as well as the formation of 2-LTR circles was notably reduced. Reporter assays showed that CD63 down regulation reduced production of the early HIV protein Tat. In agreement, CD63 silencing also inhibited production of the late protein p24. These findings suggest that CD63 plays an early post-entry role prior to or at the reverse transcription step.
Collapse
Affiliation(s)
- Guangyu Li
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | | | | | | | | |
Collapse
|
24
|
Friedrich B, Li G, Dziuba N, Ferguson MR. Quantitative PCR used to assess HIV-1 integration and 2-LTR circle formation in human macrophages, peripheral blood lymphocytes and a CD4+ cell line. Virol J 2010; 7:354. [PMID: 21129188 PMCID: PMC3003270 DOI: 10.1186/1743-422x-7-354] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 12/03/2010] [Indexed: 01/07/2023] Open
Abstract
Background Integration is an intermediate step in the HIV life cycle and is defined as the insertion of HIV-1 proviral DNA into the host chromosome. If integration does not occur when HIV-1 cDNA enters the nucleus, it circularizes upon itself and forms a 2-LTR circle. Monitoring the level of integrated HIV-1 cDNA in different primary cell subsets is very important, particularly regarding the effect of HAART in HIV-1 infected individuals. Because of limitations of prior HIV-1 integration assays, there is limited data on the level of integration and 2-LTR circle formation in primary cell subsets, particularly in human monocyte-derived macrophages and peripheral blood lymphocytes (PBL). Results In this study, we utilized a well-defined, sensitive two-step quantitative real-time PCR method to detect HIV-1 integration as well as conventional real-time PCR to detect 2-LTR circle formation in human macrophages and PBL isolated from six different healthy donors, as well as U373 CD4+ cells by infecting with HIV-1SX (R5) or dual-tropic isolate HIV-189.6 (R5/X4) virus strains. We used the FDA-approved integrase inhibitor, raltegravir, to determine quantitative differences of integrated HIV viral cDNA in HIV-1 infected cells with and without raltegravir treatment. Our results show that integration and 2-LTR circle formation can be assessed in primary macrophages, PBL, and a CD4+ cell line by this method. Specifically, our results demonstrate that this two-step real-time PCR method can distinguish between HIV-1 integrated viral cDNA and non-integrated nuclear HIV-1 2-LTR circles caused by impaired integration with raltegravir-treatment. This further confirms that only integrated HIV-1 cDNA can be specifically amplified and quantified by two-step PCR without non-specifically detecting non-integrated viral cDNA. Conclusion These results consistently demonstrate that the well-established real-time PCR assays used are robust, sensitive and quantitative for the detection of HIV-1 integration and 2-LTR circle formation in physiologically relevant human macrophages and PBL using lab-adapted virus strains, instead of pseudovirus. With two-step real-time PCR, we show that unintegrated, nuclear HIV-1 cDNA is not detected in raltegravir-treated cells, while specific for only integrated HIV-1 cDNA in non-treated cells. These methods could be applied as a useful tool in further monitoring specific therapy in HIV-1 infected individuals.
Collapse
Affiliation(s)
- Brian Friedrich
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas 77555-0435, USA
| | | | | | | |
Collapse
|
25
|
Hassuna N, Monk PN, Moseley GW, Partridge LJ. Strategies for targeting tetraspanin proteins: potential therapeutic applications in microbial infections. BioDrugs 2010; 23:341-59. [PMID: 19894777 PMCID: PMC7100176 DOI: 10.2165/11315650-000000000-00000] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The identification of novel targets and strategies for therapy of microbial infections is an area of intensive research due to the failure of conventional vaccines or antibiotics to combat both newly emerging diseases (e.g. viruses such as severe acute respiratory syndrome (SARS) and new influenza strains, and antibiotic-resistant bacteria) and entrenched, pandemic diseases exemplified by HIV. One clear approach to this problem is to target processes of the host organism rather than the microbe. Recent data have indicated that members of the tetraspanin superfamily, proteins with a widespread distribution in eukaryotic organisms and 33 members in humans, may provide such an approach. Tetraspanins traverse the membrane four times, but are distinguished from other four-pass membrane proteins by the presence of conserved charged residues in the transmembrane domains and a defining ‘signature’ motif in the larger of the two extracellular domains (the EC2). They characteristically form promiscuous associations with one another and with other membrane proteins and lipids to generate a specialized type of microdomain: the tetraspanin-enriched microdomain (TEM). TEMs are integral to the main role of tetraspanins as ‘molecular organizers’ involved in functions such as membrane trafficking, cell-cell fusion, motility, and signaling. Increasing evidence demonstrates that tetraspanins are used by intracellular pathogens as a means of entering and replicating within human cells. Although previous investigations focused mainly on viruses such as hepatitis C and HIV, it is now becoming clear that other microbes associate with tetraspanins, using TEMs as a ‘gateway’ to infection. In this article we review the properties and functions of tetraspanins/TEMs that are relevant to infective processes and discuss the accumulating evidence that shows how different pathogens exploit these properties in infection and in the pathogenesis of disease. We then investigate the novel and exciting possibilities of targeting tetraspanins for the treatment of infectious disease, using specific antibodies, recombinant EC2 domains, small-molecule mimetics, and small interfering RNA. Such therapies, directed at host-cell molecules, may provide alternative options for combating fast-mutating or newly emerging pathogens, where conventional approaches face difficulties.
Collapse
Affiliation(s)
- Noha Hassuna
- Department of Molecular Biology and Biotechnology, Krebs Institute, University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
26
|
Chambrion C, Le Naour F. The tetraspanins CD9 and CD81 regulate CD9P1-induced effects on cell migration. PLoS One 2010; 5:e11219. [PMID: 20574531 PMCID: PMC2888588 DOI: 10.1371/journal.pone.0011219] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 05/20/2010] [Indexed: 12/15/2022] Open
Abstract
CD9P-1 is a cell surface protein with immunoglobulin domains and an unknown function that specifically associates with tetraspanins CD9 and CD81. Overexpression of CD9P-1 in HEK-293 cells induces dramatic changes in cell spreading and migration on various matrices. Experiments using time-lapse videomicroscopy revealed that CD9P-1 expression has led to higher cell motility on collagen I but lower motility on fibronectin through a beta1-integrins dependent mechanism. On collagen I, the increase in cell motility induced by CD9P-1 expression was found to involve integrin alpha2beta1 and CD9P-1 was observed to associate with this collagen receptor. The generation of CD9P-1 mutants demonstrated that the transmembrane and the cytoplasmic domains are necessary for inducing effects on cell motility. On the other hand, expression of tetraspanins CD9 or CD81 was shown to reverse the effects of CD9P-1 on cell motility on collagen I or fibronectin with a concomitant association with CD9P-1. Thus, the ratio of expression levels between CD9P-1 and its tetraspanin partners can regulate cell motility.
Collapse
Affiliation(s)
- Célia Chambrion
- Inserm U1004, Villejuif, France
- Univ. Paris-Sud 11, Institut André Lwoff, Villejuif, France
| | - François Le Naour
- Univ. Paris-Sud 11, Institut André Lwoff, Villejuif, France
- Inserm U785, Villejuif, France
- * E-mail:
| |
Collapse
|
27
|
Abstract
Current antiretroviral therapy regimens can effectively suppress HIV in patients for prolonged periods of time, but do not constitute a cure, since they are incapable of eradicating viral reservoirs. It is, therefore, necessary for us to refocus on the partially understood pathogenesis of HIV, on the issue of viral persistence, and on the development of strategies for a temporally contained therapy capable of purging HIV from the body. Macrophages play a pivotal role in all three of these scenarios. This review summarizes important aspects of macrophage biology as they relate to HIV and discusses conceptual challenges for virus suppression and eradication in this cell type. We highlight a number of significant recent advances in understanding differences in HIV replication and pharmacotherapy between macrophages and CD4 T cells, as well as the role of macrophages in various aspects of the disease process and in different anatomical compartments. Finally, the importance of infected macrophages in the persistence of HIV, regarding both pathogenesis and advancement of eradication strategies, is discussed.
Collapse
Affiliation(s)
- Stephanie Venzke
- Department of Virology, University of Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| | | |
Collapse
|
28
|
Abstract
Tetraspanins are small integral membrane proteins that are known to control a variety of cellular processes, including signaling, migration and cell-cell fusion. Research over the past few years established that they are also regulators of various steps in the HIV-1 replication cycle, but the mechanisms through which these proteins either enhance or repress virus spread remain largely unknown.
Collapse
Affiliation(s)
- Markus Thali
- Department of Microbiology and Molecular Genetics, College of Medicine and CALS, University of Vermont, 318 Stafford Hall, 95 Carrigan Drive, Burlington, VT 05405-0084, USA.
| |
Collapse
|
29
|
Wang B, Li F, Xiang J, Gui L, Luo Z, Yan H. Three tetraspanins from Chinese shrimp, Fenneropenaeus chinensis, may play important roles in WSSV infection. JOURNAL OF FISH DISEASES 2010; 33:15-29. [PMID: 19943839 DOI: 10.1111/j.1365-2761.2009.01079.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Three members of the tetraspanin/TM(4)SF superfamily were cloned from Chinese shrimp, Fenneropenaeus chinensis. The deduced amino acid sequences of the three proteins have typical motifs of the tetraspanin/TM(4)SF superfamily. Phylogenetic analysis of the proteins, together with the known tetraspanins of invertebrates and vertebrates, revealed that they belong to different tetraspanin subfamilies: CD9, CD63 and tetraspanin-3. The three cloned genes of CD9, CD63 and tetraspanin-3 showed apparently different tissue distributions. The CD9 gene (FcCD9) was specifically expressed in the hepatopancreas. While for the CD63 gene (FcCD63), the highest expression was detected in nerves, epidermis and heart, with low expression in haemocytes, ovary, gill, hepatopancreas and stomach and no expression in intestine, muscle and lymphoid organ. Compared with FcCD9 and FcCD63, the tetraspanin-3 gene (FcTetraspanin-3) was more broadly expressed and its highest expression was detected in the intestine. Its expression in nerves was lower than in the intestine, but was higher than in other tissues. Expression in haemocytes, ovary and muscle was much lower than in other tissues. The expression profiles of FcCD9, FcCD63 and FcTetraspanin-3 in different tissues, including haemocytes, lymphoid organ and hepatopancreas, were compared by real-time PCR when shrimp were challenged by live white spot syndrome virus (WSSV) and heat-inactivated WSSV. All three tetraspanins were markedly up-regulated in the live WSSV-challenged shrimp tissues. The data suggested that the three cloned members of TM(4)SF superfamily in Chinese shrimp may play a key role in the route of WSSV infection.
Collapse
Affiliation(s)
- B Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | | | | | | | | | | |
Collapse
|
30
|
André M, Chambrion C, Charrin S, Soave S, Chaker J, Boucheix C, Rubinstein E, Le Naour F. In situ chemical cross-linking on living cells reveals CD9P-1 cis-oligomer at cell surface. J Proteomics 2009; 73:93-102. [PMID: 19703604 DOI: 10.1016/j.jprot.2009.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 07/08/2009] [Accepted: 08/17/2009] [Indexed: 01/23/2023]
Abstract
Tetraspanins are integral membrane proteins involved in a variety of physiological and pathological processes. They associate with each other in multimolecular complexes containing numerous membrane proteins. As a first step towards the study of the supramolecular organization of tetraspanin complexes, we have implemented a proteomic approach based on in situ protein cross-linking on living cells followed by affinity purification of tetraspanin complexes. This allowed observing the presence of high molecular weight protein complexes that were characterized as containing CD9P-1/CD315 using LC-MS/MS. Western blot analyses and the use of different tags demonstrated the presence of CD9P-1 oligomer in cis-association at cell surface. A significant amount of CD9P-1 oligomer was observed on various cell types. We have shown that CD9P-1 self-associates independently from its association with tetraspanins. However, the expression level of CD9 or CD81 that associate directly and specifically with CD9P-1, positively modulates the cross-linking efficiency of CD9P-1. Thus, tetraspanins can play a role on CD9P-1 oligomerization status.
Collapse
|
31
|
Parthasarathy V, Martin F, Higginbottom A, Murray H, Moseley GW, Read RC, Mal G, Hulme R, Monk PN, Partridge LJ. Distinct roles for tetraspanins CD9, CD63 and CD81 in the formation of multinucleated giant cells. Immunology 2009; 127:237-48. [PMID: 19489128 DOI: 10.1111/j.1365-2567.2008.02945.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Members of the tetraspanin superfamily of proteins are implicated in a variety of complex cell processes including cell fusion. However, the contribution of individual tetraspanins to these processes has proved difficult to define. Here we report the use of recombinant extracellular regions of tetraspanins to investigate the role of specific members of this family in the fusion of monocytes to form multinucleated giant cells (MGC). In contrast to their positive requirement in sperm-egg fusion, previous studies using antibodies and knockout mice have indicated a negative regulatory role for tetraspanins CD9 and CD81 in this process. In an in vitro model of fusion using human monocytes, we have confirmed observations that antibodies to CD9 and CD81 enhance MGC formation; however, in contrast to previous investigations, we found that all members of a panel of antibodies to CD63 inhibited fusion. Moreover, recombinant proteins corresponding to the large extracellular domains (EC2s) of CD63 and CD9 inhibited MGC formation, whereas the EC2s of CD81 and CD151 had no effect. The potent inhibition of fusion and binding of labelled CD63 EC2 to monocytes under fusogenic conditions suggest a direct interaction with a membrane component required for fusion. Our findings indicate that the tetraspanins CD9, CD63 and CD81 are all involved in MGC formation, but play distinct roles.
Collapse
|
32
|
Krementsov DN, Weng J, Lambelé M, Roy NH, Thali M. Tetraspanins regulate cell-to-cell transmission of HIV-1. Retrovirology 2009; 6:64. [PMID: 19602278 PMCID: PMC2714829 DOI: 10.1186/1742-4690-6-64] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 07/14/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The presence of the tetraspanins CD9, CD63, CD81 and CD82 at HIV-1 budding sites, at the virological synapse (VS), and their enrichment in HIV-1 virions has been well-documented, but it remained unclear if these proteins play a role in the late phase of the viral replication cycle. Here we used overexpression and knockdown approaches to address this question. RESULTS Neither ablation of CD9, CD63 and/or CD81, nor overexpression of these tetraspanins was found to affect the efficiency of virus release. However, confirming recently reported data, tetraspanin overexpression in virus-producing cells resulted in the release of virions with substantially reduced infectivity. We also investigated the roles of these tetraspanins in cell-to-cell transmission of HIV-1. Overexpression of CD9 and CD63 led to reduced cell-to-cell transmission of this virus. Interestingly, in knockdown experiments we found that ablation of CD63, CD9 and/or CD81 had no effect on cell-free infectivity. However, knockdown of CD81, but not CD9 and CD63, enhanced productive particle transmission to target cells, suggesting additional roles for tetraspanins in the transmission process. Finally, tetraspanins were found to be downregulated in HIV-1-infected T lymphocytes, suggesting that HIV-1 modulates the levels of these proteins in order to maximize the efficiency of its transmission within the host. CONCLUSION Altogether, these results establish an active role of tetraspanins in HIV-1 producer cells.
Collapse
Affiliation(s)
- Dimitry N Krementsov
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA.
| | | | | | | | | |
Collapse
|
33
|
Abstract
A complex relationship exists between HIV and its cellular targets. The lethal effect of HIV on circulating CD4(+) helper T lymphocytes parallels the degree of the infected individual's immunodeficiency and ultimately the transition to AIDS and death. However, as with other members of the Lentivirus family of retroviruses, the ubiquitous, mobile macrophage is also a prime target for HIV infection, and apparently, in most instances, is the initial infected cell, since most people are infected with a CCR5 chemokine-tropic virus. Unlike the lymphocyte, the macrophage is apparently a more stable viral host, capable of a long infected life as an HIV reservoir and a chronic source of infectious virus. Published in vitro studies have indicated that whereas lymphocytes replicate HIV solely on their plasma membrane, macrophages have been envisaged to predominantly replicate HIV within cytoplasmic vacuoles, and thus have been likened to a "Trojan horse," when it comes to the immune system. Recent studies have revealed an ingenious way by which the cultured monocyte-derived macrophage (MDM) replicates HIV and releases it into the medium. The key macrophage organelle appears to be what is alternatively referred to as the "late endosome" (LE) or the "multivesicular body" (MVB), which have a short and a long history, respectively. Proof of the association is that chemically, LE/MVB and their vesicles possess several pathopneumonic membrane markers (e.g., CD63) that are found on released HIV particles. The hypothesis is that HIV usurps this vesicle-forming mechanism and employs it for its own replication. Release of the intravacuolar virus from the cell is hypothesized to occur by a process referred to as exocytosis, resulting from the fusion of virus-laden LE/MVB with the plasma membrane of the macrophage. Interestingly, LE/MVB are also involved in the infection stage of MDM by HIV. Close review of the literature reveals that along with the Golgi, which contributes to the formation of LE/MVB, the MVB was first identified as a site of HIV replication by macrophages many years ago, but the full implication of this observation was not appreciated at the time. As in many other areas of HIV research, what has been totally lacking is an in vivo confirmation of the in vitro phenomenon. Herein, the ultrastructure of HIV interaction with cells in vitro and in vivo is explored. It is shown that while HIV is regularly found in LE/MVB in vitro, it is infrequently the case in vivo. Therefore, the results challenge the "Trojan horse" concept.
Collapse
Affiliation(s)
- Jan Marc Orenstein
- Department of Pathology, George Washington University Medical Center, Washington, DC 20037-2336, USA.
| |
Collapse
|
34
|
Formation of syncytia is repressed by tetraspanins in human immunodeficiency virus type 1-producing cells. J Virol 2009; 83:7467-74. [PMID: 19458002 DOI: 10.1128/jvi.00163-09] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In vitro propagation studies have established that human immunodeficiency virus type 1 (HIV-1) is most efficiently transmitted at the virological synapse that forms between producer and target cells. Despite the presence of the viral envelope glycoprotein (Env) and CD4 and chemokine receptors at the respective surfaces, producer and target cells usually do not fuse with each other but disengage after the viral particles have been delivered, consistent with the idea that syncytia, at least in vitro, are not required for HIV-1 spread. Here, we tested whether tetraspanins, which are well known regulators of cellular membrane fusion processes that are enriched at HIV-1 exit sites, regulate syncytium formation. We found that overexpression of tetraspanins in producer cells leads to reduced syncytium formation, while downregulation has the opposite effect. Further, we document that repression of Env-induced cell-cell fusion by tetraspanins depends on the presence of viral Gag, and we demonstrate that fusion repression requires the recruitment of Env by Gag to tetraspanin-enriched microdomains (TEMs). However, sensitivity to fusion repression by tetraspanins varied for different viral strains, despite comparable recruitment of their Envs to TEMs. Overall, these data establish tetraspanins as negative regulators of HIV-1-induced cell-cell fusion, and they start delineating the requirements for this regulation.
Collapse
|
35
|
Abstract
Despite high expression levels at the plasma membrane or in intracellular vesicles, tetraspanins remain among the most mysterious transmembrane molecules 20 years after their discovery. Several genetic studies in mammals and invertebrates have demonstrated key physiological roles for some of these tetraspanins, in particular in the immune response, sperm-egg fusion, photoreceptor function and the normal function of certain epithelia. Other studies have highlighted their ability to modulate cell migration and metastasis formation. Their role in the propagation of infectious agents has drawn recent attention, with evidence for HIV budding in tetraspanin-enriched plasma membrane domains. Infection of hepatocytic cells by two major pathogens, the hepatitis C virus and the malaria parasite, also requires the tetraspanin CD81. The function of tetraspanins is thought to be linked to their ability to associate with one another and a wealth of other integral proteins, thereby building up an interacting network or 'tetraspanin web'. On the basis of the biochemical dissection of the tetraspanin web and recent analysis of the dynamics of some of its constituents, we propose that tetraspanins tightly regulate transient interactions between a variety of molecules and as such favour the efficient assembly of specialized structures upon proper stimulation.
Collapse
|
36
|
Hemler ME. Targeting of tetraspanin proteins--potential benefits and strategies. Nat Rev Drug Discov 2009; 7:747-58. [PMID: 18758472 DOI: 10.1038/nrd2659] [Citation(s) in RCA: 268] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tetraspanin transmembrane proteins have emerged as key players in malignancy, the immune system, during fertilization and infectious disease processes. Tetraspanins engage in a wide range of specific molecular interactions, occurring through the formation of tetraspanin-enriched microdomains (TEMs). TEMs therefore serve as a starting point for understanding how tetraspanins affect cell signalling, adhesion, morphology, motility, fusion and virus infection. An abundance of recent evidence suggests that targeting tetraspanins, for example, by monoclonal antibodies, soluble large-loop proteins or RNAi technology, should be therapeutically beneficial.
Collapse
Affiliation(s)
- Martin E Hemler
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, Massachussetts 02115, USA.
| |
Collapse
|
37
|
Fumagalli M, Cagliani R, Pozzoli U, Riva S, Comi GP, Menozzi G, Bresolin N, Sironi M. Widespread balancing selection and pathogen-driven selection at blood group antigen genes. Genes Dev 2009; 19:199-212. [PMID: 18997004 PMCID: PMC2652214 DOI: 10.1101/gr.082768.108] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 11/04/2008] [Indexed: 12/31/2022]
Abstract
Historically, allelic variations in blood group antigen (BGA) genes have been regarded as possible susceptibility factors for infectious diseases. Since host-pathogen interactions are major determinants in evolution, BGAs can be thought of as selection targets. In order to verify this hypothesis, we obtained an estimate of pathogen richness for geographic locations corresponding to 52 populations distributed worldwide; after correction for multiple tests and for variables different from selective forces, significant correlations with pathogen richness were obtained for multiple variants at 11 BGA loci out of 26. In line with this finding, we demonstrate that three BGA genes, namely CD55, CD151, and SLC14A1, have been subjected to balancing selection, a process, rare outside MHC genes, which maintains variability at a locus. Moreover, we identified a gene region immediately upstream the transcription start site of FUT2 which has undergone non-neutral evolution independently from the coding region. Finally, in the case of BSG, we describe the presence of a highly divergent haplotype clade and the possible reasons for its maintenance, including frequency-dependent balancing selection, are discussed. These data indicate that BGAs have been playing a central role in the host-pathogen arms race during human evolutionary history and no other gene category shows similar levels of widespread selection, with the only exception of loci involved in antigen recognition.
Collapse
Affiliation(s)
- Matteo Fumagalli
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, 23842 Bosisio Parini (LC), Italy
- Bioengineering Department, Politecnico di Milano, 20133 Milan, Italy
| | - Rachele Cagliani
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, 23842 Bosisio Parini (LC), Italy
| | - Uberto Pozzoli
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, 23842 Bosisio Parini (LC), Italy
| | - Stefania Riva
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, 23842 Bosisio Parini (LC), Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Department of Neurological Sciences, University of Milan, IRCCS Ospedale Maggiore Policlinico, Mangiagalli and Regina Elena Foundation, 20100 Milan, Italy
| | - Giorgia Menozzi
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, 23842 Bosisio Parini (LC), Italy
| | - Nereo Bresolin
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, 23842 Bosisio Parini (LC), Italy
- Dino Ferrari Centre, Department of Neurological Sciences, University of Milan, IRCCS Ospedale Maggiore Policlinico, Mangiagalli and Regina Elena Foundation, 20100 Milan, Italy
| | - Manuela Sironi
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, 23842 Bosisio Parini (LC), Italy
| |
Collapse
|
38
|
Deficiency of the tetraspanin CD63 associated with kidney pathology but normal lysosomal function. Mol Cell Biol 2008; 29:1083-94. [PMID: 19075008 DOI: 10.1128/mcb.01163-08] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD63 is a member of the tetraspanin superfamily that constitutes a main component of the lysosomal membrane. In mice, two CD63 gene loci are present, with only one of these two being functional. We generated and analyzed mice deficient for active CD63. Disruption of CD63 results in a complete loss of CD63 protein expression. Despite its abundance in late endosomes/lysosomes, the lack of CD63 does not cause obvious endosomal/lysosomal abnormalities. CD63 knockout mice are viable and fertile without gross morphological abnormalities in the majority of tissues. No alterations in the populations of immune cells and only minor differences in platelet function were observed. This suggests that the lack of CD63 could be successfully compensated for, most likely by other tetraspanins. However, CD63 deficiency leads to an altered water balance. CD63 knockout mice show an increased urinary flow, water intake, reduced urine osmolality, and a higher fecal water content. In principle cells of the collecting duct of CD63-deficient mice, abnormal intracellular lamellar inclusions were observed. This indicates that the sorting of apical transport proteins might be impaired in these cells. CD63 knockout mice provide an important tool for analyzing the various postulated functions of CD63 in vivo.
Collapse
|
39
|
Spoden G, Freitag K, Husmann M, Boller K, Sapp M, Lambert C, Florin L. Clathrin- and caveolin-independent entry of human papillomavirus type 16--involvement of tetraspanin-enriched microdomains (TEMs). PLoS One 2008; 3:e3313. [PMID: 18836553 PMCID: PMC2561052 DOI: 10.1371/journal.pone.0003313] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Accepted: 09/10/2008] [Indexed: 11/19/2022] Open
Abstract
Background Infectious entry of human papillomaviruses into their host cells is an important step in the viral life cycle. For cell binding these viruses use proteoglycans as initial attachment sites. Subsequent transfer to a secondary receptor molecule seems to be involved in virus uptake. Depending on the papillomavirus subtype, it has been reported that entry occurs by clathrin- or caveolin-mediated mechanisms. Regarding human papillomavirus type 16 (HPV16), the primary etiologic agent for development of cervical cancer, clathrin-mediated endocytosis was described as infectious entry pathway. Methodology/Principal Findings Using immunofluorescence and infection studies we show in contrast to published data that infectious entry of HPV16 occurs in a clathrin- and caveolin-independent manner. Inhibition of clathrin- and caveolin/raft-dependent endocytic pathways by dominant-negative mutants and siRNA-mediated knockdown, as well as inhibition of dynamin function, did not impair infection. Rather, we provide evidence for involvement of tetraspanin-enriched microdomains (TEMs) in HPV16 endocytosis. Following cell attachment, HPV16 particles colocalized with the tetraspanins CD63 and CD151 on the cell surface. Notably, tetraspanin-specific antibodies and siRNA inhibited HPV16 cell entry and infection, confirming the importance of TEMs for infectious endocytosis of HPV16. Conclusions/Significance Tetraspanins fulfill various roles in the life cycle of a number of important viral pathogens, including human immunodeficiency virus (HIV) and hepatitis C virus (HCV). However, their involvement in endocytosis of viral particles has not been proven. Our data indicate TEMs as a novel clathrin- and caveolin-independent invasion route for viral pathogens and especially HPV16.
Collapse
Affiliation(s)
- Gilles Spoden
- Institute for Medical Microbiology and Hygiene, University of Mainz, Mainz, Germany
| | - Kirsten Freitag
- Institute for Medical Microbiology and Hygiene, University of Mainz, Mainz, Germany
| | - Matthias Husmann
- Institute for Medical Microbiology and Hygiene, University of Mainz, Mainz, Germany
| | | | - Martin Sapp
- Department of Microbiology and Immunology, Feist-Weiller Cancer Center, Center for Molecular Tumor Virology, LSU Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Carsten Lambert
- Institute for Medical Microbiology and Hygiene, University of Mainz, Mainz, Germany
- * E-mail:
| | - Luise Florin
- Institute for Medical Microbiology and Hygiene, University of Mainz, Mainz, Germany
| |
Collapse
|
40
|
Chen H, Dziuba N, Friedrich B, von Lindern J, Murray JL, Rojo DR, Hodge TW, O'Brien WA, Ferguson MR. A critical role for CD63 in HIV replication and infection of macrophages and cell lines. Virology 2008; 379:191-6. [PMID: 18682304 DOI: 10.1016/j.virol.2008.06.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 02/15/2008] [Accepted: 06/24/2008] [Indexed: 11/25/2022]
Abstract
HIV infection typically involves interaction of Env with CD4 and a chemokine coreceptor, either CCR5 or CXCR4. Other cellular factors supporting HIV replication have also been characterized. We previously demonstrated a role for CD63 in early HIV infection events in macrophages via inhibition by anti-CD63 antibody pretreatment. To confirm the requirement for CD63 in HIV replication, we decreased CD63 expression using CD63-specific short interfering RNAs (siRNA), and showed inhibition of HIV replication in macrophages. Surprisingly, pretreatment with CD63 siRNA not only silenced CD63 expression by 90%, but also inhibited HIV-1 replication in a cultured cell line (U373-MAGI) which had been previously shown to be insensitive to CD63 monoclonal antibody inhibition. Although the anti-CD63 antibody was previously shown to inhibit early HIV infection events only in macrophages, we now show a potential role for CD63 in later HIV replication events in macrophages and cell lines. Further delineation of the role of CD63 in HIV replication may lead to development of novel therapeutic compounds.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yoshida T, Kawano Y, Sato K, Ando Y, Aoki J, Miura Y, Komano J, Tanaka Y, Koyanagi Y. A CD63 Mutant Inhibits T-cell Tropic Human Immunodeficiency Virus Type 1 Entry by Disrupting CXCR4 Trafficking to the Plasma Membrane. Traffic 2008; 9:540-58. [DOI: 10.1111/j.1600-0854.2007.00700.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
42
|
CD63 is not required for production of infectious human immunodeficiency virus type 1 in human macrophages. J Virol 2008; 82:4751-61. [PMID: 18321974 DOI: 10.1128/jvi.02320-07] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During the assembly of human immunodeficiency virus type 1 (HIV-1) particles, the tetraspanin CD63 can be incorporated into the viral membrane. Indeed, cell surface tetraspanin microdomains that include CD63 have been proposed as sites for virus release. In addition, antibodies against CD63 can inhibit HIV infection of macrophages. In this cell type, HIV assembles into intracellularly sequestered plasma membrane domains that contain several other tetraspanins, including CD81, CD9, and CD53. CD63 is recruited to this domain following HIV infection. Together, these observations suggest that CD63 may have some function in the assembly of infectious virus particles and/or the infectivity of assembled virions. Here we have used RNA interference to knock down CD63 expression in monocyte-derived primary macrophages. We show that in the absence of CD63, HIV assembly is quantitatively comparable to that seen in CD63-expressing macrophages and that virus assembly occurs on compartments positive for CD81, CD9, and CD53. Moreover, the infectivity of macrophage-derived virus is unaffected by the loss of CD63. Together, our results indicate that at least in tissue culture, CD63 expression is not required for either the production or the infectivity of HIV-1.
Collapse
|
43
|
Abache T, Le Naour F, Planchon S, Harper F, Boucheix C, Rubinstein E. The transferrin receptor and the tetraspanin web molecules CD9, CD81, and CD9P-1 are differentially sorted into exosomes after TPA treatment of K562 cells. J Cell Biochem 2008; 102:650-64. [PMID: 17407154 DOI: 10.1002/jcb.21318] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Here we show that treatment of K562 cells with the phorbol ester TPA induces the down-modulation of various surface antigens. Among them, the transferrin receptor (TfR), the tetraspanin CD81, and a CD81-associated protein, CD9P-1, were unique in that their expression levels were lower after 24 h incubation than after 3 h. We demonstrated that like the TfR, CD81 was internalized at early times, and was less synthesized at latter times. Despite the association of a fraction of the TfR with CD81, these two molecules were subjected to different fates. TPA increased targeting of CD81 and CD9P-1 into exosomes but strongly reduced the localization of the TfR in these vesicles. Using this model we have shown that a fraction of CD81 and CD9P-1 in exosomes comes from a surface pool and that these molecules remain associated in exosomes. However, CD9P-1 could be targeted to exosomes in the absence of CD81 and of another tetraspanin, CD9. The targeting of CD9 into exosomes did not require palmitoylation of the protein. J. Cell. Biochem. 102: 650-664, 2007. (c) 2007 Wiley-Liss, Inc.
Collapse
|
44
|
Modulation of human immunodeficiency virus type 1 infectivity through incorporation of tetraspanin proteins. J Virol 2007; 82:1021-33. [PMID: 17989173 DOI: 10.1128/jvi.01044-07] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Accumulating evidence indicates that human immunodeficiency virus type 1 (HIV-1) acquires various cellular membrane proteins in the lipid bilayer of the viral envelope membrane. Although some virion-incorporated cellular membrane proteins are known to potently affect HIV-1 infectivity, the virological functions of most virion-incorporated membrane proteins remain unclear. Among these host proteins, we found that CD63 was eliminated from the plasma membranes of HIV-1-producing T cells after activation, followed by a decrease in the amount of virion-incorporated CD63, and in contrast, an increase in the infectivity of the released virions. On the other hand, we found that CD63 at the cell surface was preferentially embedded on the membrane of released virions in an HIV-1 envelope protein (Env)-independent manner and that virion-incorporated CD63 had the potential to inhibit HIV-1 Env-mediated infection in a strain-specific manner at the postattachment entry step(s). In addition, these behaviors were commonly observed in other tetraspanin proteins, such as CD9, CD81, CD82, and CD231. However, L6 protein, whose topology is similar to that of tetraspanins but which does not belong to the tetraspanin superfamily, did not have the potential to prevent HIV-1 infection, despite its successful incorporation into the released particles. Taken together, these results suggest that tetraspanin proteins have the unique potential to modulate HIV-1 infectivity through incorporation into released HIV-1 particles, and our findings may provide a clue to undiscovered aspects of HIV-1 entry.
Collapse
|
45
|
André M, Morelle W, Planchon S, Milhiet PE, Rubinstein E, Mollicone R, Chamot-Rooke J, Le Naour F. Glycosylation status of the membrane protein CD9P-1. Proteomics 2007; 7:3880-95. [DOI: 10.1002/pmic.200700355] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
46
|
WFDC1/ps20 is a novel innate immunomodulatory signature protein of human immunodeficiency virus (HIV)-permissive CD4+ CD45RO+ memory T cells that promotes infection by upregulating CD54 integrin expression and is elevated in HIV type 1 infection. J Virol 2007; 82:471-86. [PMID: 17942534 DOI: 10.1128/jvi.00939-07] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Understanding why human immunodeficiency virus (HIV) preferentially infects some CD4(+) CD45RO(+) memory T cells has implications for antiviral immunity and pathogenesis. We report that differential expression of a novel secreted factor, ps20, previously implicated in tissue remodeling, may underlie why some CD4 T cells are preferentially targeted. We show that (i) there is a significant positive correlation between endogenous ps20 mRNA in diverse CD4 T-cell populations and in vitro infection, (ii) a ps20(+) permissive cell can be made less permissive by antibody blockade- or small-interference RNA-mediated knockdown of endogenous ps20, and (iii) conversely, a ps20(low) cell can be more permissive by adding ps20 exogenously or engineering stable ps20 expression by retroviral transduction. ps20 expression is normally detectable in CD4 T cells after in vitro activation and interleukin-2 expansion, and such oligoclonal populations comprise ps20(positive) and ps20(low/negative) isogenic clones at an early differentiation stage (CD45RO(+)/CD25(+)/CD28(+)/CD57(-)). This pattern is altered in chronic HIV infection, where ex vivo CD4(+) CD45RO(+) T cells express elevated ps20. ps20 promoted HIV entry via fusion and augmented CD54 integrin expression; both of these effects were reversed by anti-ps20 antibody. We therefore propose ps20 to be a novel signature of HIV-permissive CD4 T cells that promotes infection in an autocrine and paracrine manner and that HIV has coopted a fundamental role of ps20 in promoting cell adhesion for its benefit. Disrupting the ps20 pathway may therefore provide a novel anti-HIV strategy.
Collapse
|
47
|
Khurana S, Krementsov DN, de Parseval A, Elder JH, Foti M, Thali M. Human immunodeficiency virus type 1 and influenza virus exit via different membrane microdomains. J Virol 2007; 81:12630-40. [PMID: 17855546 PMCID: PMC2168970 DOI: 10.1128/jvi.01255-07] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Directed release of human immunodeficiency virus type 1 (HIV-1) into the cleft of the virological synapse that can form between infected and uninfected T cells, for example, in lymph nodes, is thought to contribute to the systemic spread of this virus. In contrast, influenza virus, which causes local infections, is shed into the airways of the respiratory tract from free surfaces of epithelial cells. We now demonstrate that such differential release of HIV-1 and influenza virus is paralleled, at the subcellular level, by viral assembly at different microsegments of the plasma membrane of HeLa cells. HIV-1, but not influenza virus, buds through microdomains containing the tetraspanins CD9 and CD63. Consequently, the anti-CD9 antibody K41, which redistributes its antigen and also other tetraspanins to cell-cell adhesion sites, interferes with HIV-1 but not with influenza virus release. Altogether, these data strongly suggest that the bimodal egress of these two pathogenic viruses, like their entry into target cells, is guided by specific sets of host cell proteins.
Collapse
Affiliation(s)
- Sandhya Khurana
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | |
Collapse
|
48
|
Pan-Hammarström Q, Wen S, Hammarström L. Cytokine gene expression profiles in human lymphocytes induced by a formula of traditional Chinese medicine, vigconic VI-28. J Interferon Cytokine Res 2007; 26:628-36. [PMID: 16978066 DOI: 10.1089/jir.2006.26.628] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
VI-28 is a formula of traditional Chinese medicine (TCM) that has been used in aged individuals to improve health and, recently, to treat patients with chronic human immunodeficiency virus (HIV) and hepatitis B virus (HBV) infections. The mechanism underlying its clinical effect is, however, largely unknown. In the current study, we used a transwell culture system that mimics the in vivo situation and applied microarray technology to explore the effect of VI-28 on gene expression in human lymphocytes. The VI-28 treatment induced expression of a number of proinflammatory cytokines/chemokines in both peripheral blood mononuclear cells (PBMC) and spleen cells, including interleukin-1 (IL-1), growth-related protein-beta (GRO-beta) and epithelial cellderived neutrophil-activating peptide (ENA-78 [CXCL5]). Furthermore, a specific upregulation of interferon- gamma (IFN-gamma), monokine induced by gamma interferon (MIG [CXCL9]) and interleukin-2 receptor alpha (IL-2Ralpha) in spleen cells was noted, whereas tissue inhibitor of metalloproteinase-3 (TIMP-3) and disabled-2 (DAB2) were downregulated. VI-28 might, thus, enhance both innate and acquired immunity, in particular, T cell function. In addition, genes with no obvious immunologic function, such as insulin-like growth factor-2 (IGF- 1) and CD9, were also differentially affected. Further analysis of individual and combination of ingredients of VI-28 may shed light on the role of this herbal medicine in combating different diseases.
Collapse
Affiliation(s)
- Qiang Pan-Hammarström
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden.
| | | | | |
Collapse
|
49
|
Gordón-Alonso M, Yañez-Mó M, Barreiro O, Alvarez S, Muñoz-Fernández MA, Valenzuela-Fernández A, Sánchez-Madrid F. Tetraspanins CD9 and CD81 modulate HIV-1-induced membrane fusion. THE JOURNAL OF IMMUNOLOGY 2007; 177:5129-37. [PMID: 17015697 DOI: 10.4049/jimmunol.177.8.5129] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Protein organization on the membrane of target cells may modulate HIV-1 transmission. Since the tetraspanin CD81 is associated to CD4, the receptor of HIV-1 envelope protein (Env; gp120/gp41), we have explored the possibility that this molecule may modulate the initial steps of HIV-1 infection. On the other hand, CD81 belongs to the tetraspanin family, which has been described as organizers of protein microdomains on the plasma membrane. Therefore, the role of CD81 and other related tetraspanin, CD9, on the cell-to-cell fusion process mediated by HIV-1 was studied. We found that anti-tetraspanin Abs enhanced the syncytia formation induced by HIV-1 envelope proteins and viral entry in human T lymphoblasts. In addition, anti-CD81 Abs triggered its clustering in patches, where CD4 and CXCR4 were included. Moreover, the knocking down of CD81 and CD9 expression resulted in an increase in syncytia formation and viral entry. Accordingly, overexpression of CD81 and CD9 rendered cells less susceptible to Env-mediated syncytia formation. These data indicate that CD9 and CD81 have an important role in membrane fusion induced by HIV-1 envelope.
Collapse
Affiliation(s)
- Mónica Gordón-Alonso
- Servicio de Inmunología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
50
|
Wahl SM, Greenwell-Wild T, Vázquez N. HIV accomplices and adversaries in macrophage infection. J Leukoc Biol 2006; 80:973-83. [PMID: 16908514 DOI: 10.1189/jlb.0306130] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cell surface and intracellular proteins in macrophages influence various steps in the life cycle of lentiviruses. Characterization of these restriction and/or cofactors is essential to understanding how macrophages become unwitting HIV hosts and in fact, can coexist with a heavy viral burden. Although many of the cellular pathways co-opted by HIV in macrophages mimic those seen in CD4+ T cells, emerging evidence reveals cellular constituents of the macrophage, which may be uniquely usurped by HIV. For example, in addition to CD4 and CCR5, membrane annexin II facilitates early steps in infection of macrophages, but not in T cells. Blockade of this pathway effectively diminishes macrophage infection. Viral binding engages a macrophage-centric signaling pathway and a transcriptional profile, including genes such as p21, which benefit the virus. Once inside the cell, multiple host cell molecules are engaged to facilitate virus replication and assembly. Although the macrophage is an enabler, it also possesses innate antiviral mechanisms, including apolipoprotein B mRNA-editing enzyme-catalytic polypeptide-like 3G (APOBEC3) family DNA-editing enzymes to inhibit replication of HIV. Differential expression of these enzymes, which are largely neutralized by HIV to protect its rebirth, is associated with resistance or susceptibility to the virus. Higher levels of the cytidine deaminases endow potential HIV targets with a viral shield, and IFN-alpha, a natural inducer of macrophage APOBEC expression, renders macrophages tougher combatants to HIV infection. These and other manipulatable pathways may give the macrophage a fighting chance in its battle against the virus.
Collapse
Affiliation(s)
- Sharon M Wahl
- Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Rm. 320, 30 Convent Dr., MSC 4352, Bethesda, MD 20892-4352, USA.
| | | | | |
Collapse
|