1
|
Stitz J. Development of HIV-1 vectors pseudotyped with envelope proteins of other retroviruses. Virology 2025; 602:110300. [PMID: 39577275 DOI: 10.1016/j.virol.2024.110300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/25/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
In the past three decades, human immunodeficiency virus type 1 (HIV-1)-derived vectors were evolved and became indispensable to transduce therapeutic genes into a range of different target cell types to facilitate a variety of gene therapeutic strategies. To achieve this, i) the biosafety profile of the vectors was incrementally enhanced and ii) the CD4-restricted tropism mediated by the envelope proteins (Env) of the parental virus needed to be directed towards recruitment of other receptors expressed on the desired target cells. Here, a closer look is first taken at the development of vector components and the mechanisms of Env incorporation into particles. While envelope proteins originating from a broad range of very diverse virus species were successfully utilized, members of the Retroviridae family most frequently provided Env or further engineered variants thereof to form transduction-competent HIV-1 pseudotype vector particles. The development of these vectors is reviewed and anticipated to further contribute to the future progression of somatic gene therapy.
Collapse
Affiliation(s)
- Jörn Stitz
- Research Group Medical Biotechnology & Bioengineering, Faculty of Applied Natural Sciences, TH Köln - University of Applied Sciences, Campusplatz 1, 51379, Leverkusen, Germany.
| |
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
|
Muecksch F, Klaus S, Laketa V, Müller B, Kräusslich HG. Probing Gag-Env dynamics at HIV-1 assembly sites using live-cell microscopy. J Virol 2024; 98:e0064924. [PMID: 39136462 PMCID: PMC11406925 DOI: 10.1128/jvi.00649-24] [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: 04/09/2024] [Accepted: 07/02/2024] [Indexed: 09/18/2024] Open
Abstract
Human immunodeficiency virus (HIV)-1 assembly is initiated by Gag binding to the inner leaflet of the plasma membrane (PM). Gag targeting is mediated by its N-terminally myristoylated matrix (MA) domain and PM phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Upon Gag assembly, envelope (Env) glycoproteins are recruited to assembly sites; this process depends on the MA domain of Gag and the Env cytoplasmic tail. To investigate the dynamics of Env recruitment, we applied a chemical dimerizer system to manipulate HIV-1 assembly by reversible PI(4,5)P2 depletion in combination with super resolution and live-cell microscopy. This approach enabled us to control and synchronize HIV-1 assembly and track Env recruitment to individual nascent assembly sites in real time. Single virion tracking revealed that Gag and Env are accumulating at HIV-1 assembly sites with similar kinetics. PI(4,5)P2 depletion prevented Gag PM targeting and Env cluster formation, confirming Gag dependence of Env recruitment. In cells displaying pre-assembled Gag lattices, PI(4,5)P2 depletion resulted in the disintegration of the complete assembly domain, as not only Gag but also Env clusters were rapidly lost from the PM. These results argue for the existence of a Gag-induced and -maintained membrane micro-environment, which attracts Env. Gag cluster dissociation by PI(4,5)P2 depletion apparently disrupts this micro-environment, resulting in the loss of Env from the former assembly domain.IMPORTANCEHuman immunodeficiency virus (HIV)-1 assembles at the plasma membrane of infected cells, resulting in the budding of membrane-enveloped virions. HIV-1 assembly is a complex process initiated by the main structural protein of HIV-1, Gag. Interestingly, HIV-1 incorporates only a few envelope (Env) glycoproteins into budding virions, although large Env accumulations surrounding nascent Gag assemblies are detected at the plasma membrane of HIV-expressing cells. The matrix domain of Gag and the Env cytoplasmatic tail play a role in Env recruitment to HIV-1 assembly sites and its incorporation into nascent virions. However, the regulation of these processes is incompletely understood. By combining a chemical dimerizer system to manipulate HIV-1 assembly with super resolution and live-cell microscopy, our study provides new insights into the interplay between Gag, Env, and host cell membranes during viral assembly and into Env incorporation into HIV-1 virions.
Collapse
Affiliation(s)
- Frauke Muecksch
- Department of Infectious Diseases, Virology, Heidelberg University Medical Faculty, Center for Infectious Diseases Research (CIID), Heidelberg, Germany
- Chica and Heinz Schaller (CHS) Research Group, Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany
| | - Severina Klaus
- Department of Infectious Diseases, Virology, Heidelberg University Medical Faculty, Center for Infectious Diseases Research (CIID), Heidelberg, Germany
| | - Vibor Laketa
- Department of Infectious Diseases, Virology, Heidelberg University Medical Faculty, Center for Infectious Diseases Research (CIID), Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Barbara Müller
- Department of Infectious Diseases, Virology, Heidelberg University Medical Faculty, Center for Infectious Diseases Research (CIID), Heidelberg, Germany
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Heidelberg University Medical Faculty, Center for Infectious Diseases Research (CIID), Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
Ying Y, Yang Y, Chen AK. Roles of RNA scaffolding in nanoscale Gag multimerization and selective protein sorting at HIV membranes. SCIENCE ADVANCES 2024; 10:eadk8297. [PMID: 38394201 PMCID: PMC10889351 DOI: 10.1126/sciadv.adk8297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
HIV-1 Gag proteins can multimerize upon the viral genomic RNA or multiple random cellular messenger RNAs to form a virus particle or a virus-like particle, respectively. To date, whether the two types of particles form via the same Gag multimerization process has remained unclarified. Using photoactivated localization microscopy to illuminate Gag organizations and dynamics at the nanoscale, here, we showed that genomic RNA mediates Gag multimerization in a more cluster-centric, cooperative, and spatiotemporally coordinated fashion, with the ability to drive dense Gag clustering dependent on its ability to act as a long-stranded scaffold not easily attainable by cellular messenger RNAs. These differences in Gag multimerization were further shown to affect downstream selective protein sorting into HIV membranes, indicating that the choice of RNA for packaging can modulate viral membrane compositions. These findings should advance the understanding of HIV assembly and further benefit the development of virus-like particle-based therapeutics.
Collapse
Affiliation(s)
- Yachen Ying
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Yantao Yang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Antony K Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
5
|
Thomas B, Chockalingam K, Chen Z. Methods for Engineering Binders to Multi-Pass Membrane Proteins. Bioengineering (Basel) 2023; 10:1351. [PMID: 38135942 PMCID: PMC10741020 DOI: 10.3390/bioengineering10121351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/11/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Numerous potential drug targets, including G-protein-coupled receptors and ion channel proteins, reside on the cell surface as multi-pass membrane proteins. Unfortunately, despite advances in engineering technologies, engineering biologics against multi-pass membrane proteins remains a formidable task. In this review, we focus on the different methods used to prepare/present multi-pass transmembrane proteins for engineering target-specific biologics such as antibodies, nanobodies and synthetic scaffold proteins. The engineered biologics exhibit high specificity and affinity, and have broad applications as therapeutics, probes for cell staining and chaperones for promoting protein crystallization. We primarily cover publications on this topic from the past 10 years, with a focus on the different formats of multi-pass transmembrane proteins. Finally, the remaining challenges facing this field and new technologies developed to overcome a number of obstacles are discussed.
Collapse
Affiliation(s)
- Benjamin Thomas
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX 77845, USA;
| | - Karuppiah Chockalingam
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
| | - Zhilei Chen
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX 77845, USA;
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
| |
Collapse
|
6
|
Tomishige N, Bin Nasim M, Murate M, Pollet B, Didier P, Godet J, Richert L, Sako Y, Mély Y, Kobayashi T. HIV-1 Gag targeting to the plasma membrane reorganizes sphingomyelin-rich and cholesterol-rich lipid domains. Nat Commun 2023; 14:7353. [PMID: 37990014 PMCID: PMC10663554 DOI: 10.1038/s41467-023-42994-w] [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: 08/30/2021] [Accepted: 10/26/2023] [Indexed: 11/23/2023] Open
Abstract
Although the human immunodeficiency virus type 1 lipid envelope has been reported to be enriched with host cell sphingomyelin and cholesterol, the molecular mechanism of the enrichment is not well understood. Viral Gag protein plays a central role in virus budding. Here, we report the interaction between Gag and host cell lipids using different quantitative and super-resolution microscopy techniques in combination with specific probes that bind endogenous sphingomyelin and cholesterol. Our results indicate that Gag in the inner leaflet of the plasma membrane colocalizes with the outer leaflet sphingomyelin-rich domains and cholesterol-rich domains, enlarges sphingomyelin-rich domains, and strongly restricts the mobility of sphingomyelin-rich domains. Moreover, Gag multimerization induces sphingomyelin-rich and cholesterol-rich lipid domains to be in close proximity in a curvature-dependent manner. Our study suggests that Gag binds, coalesces, and reorganizes pre-existing lipid domains during assembly.
Collapse
Affiliation(s)
- Nario Tomishige
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.
- Cellular Informatics Laboratory, RIKEN CPR, Wako, Saitama, Japan.
| | - Maaz Bin Nasim
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Motohide Murate
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
- Cellular Informatics Laboratory, RIKEN CPR, Wako, Saitama, Japan
| | - Brigitte Pollet
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Pascal Didier
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Julien Godet
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Ludovic Richert
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN CPR, Wako, Saitama, Japan
| | - Yves Mély
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.
| | - Toshihide Kobayashi
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.
- Cellular Informatics Laboratory, RIKEN CPR, Wako, Saitama, Japan.
| |
Collapse
|
7
|
Iida N, Kawahara M, Hirota R, Shibagaki Y, Hattori S, Morikawa Y. A Proteomic Analysis of Detergent-Resistant Membranes in HIV Virological Synapse: The Involvement of Vimentin in CD4 Polarization. Viruses 2023; 15:1266. [PMID: 37376566 DOI: 10.3390/v15061266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The cell-cell contact between HIV-1-infected and uninfected cells forms a virological synapse (VS) to allow for efficient HIV-1 transmission. Not only are HIV-1 components polarized and accumulate at cell-cell interfaces, but viral receptors and lipid raft markers are also. To better understand the nature of the HIV-1 VS, detergent-resistant membrane (DRM) fractions were isolated from an infected-uninfected cell coculture and compared to those from non-coculture samples using 2D fluorescence difference gel electrophoresis. Mass spectrometry revealed that ATP-related enzymes (ATP synthase subunit and vacuolar-type proton ATPase), protein translation factors (eukaryotic initiation factor 4A and mitochondrial elongation factor Tu), protein quality-control-related factors (protein disulfide isomerase A3 and 26S protease regulatory subunit), charged multivesicular body protein 4B, and vimentin were recruited to the VS. Membrane flotation centrifugation of the DRM fractions and confocal microscopy confirmed these findings. We further explored how vimentin contributes to the HIV-1 VS and found that vimentin supports HIV-1 transmission through the recruitment of CD4 to the cell-cell interface. Since many of the molecules identified in this study have previously been suggested to be involved in HIV-1 infection, we suggest that a 2D difference gel analysis of DRM-associated proteins may reveal the molecules that play crucial roles in HIV-1 cell-cell transmission.
Collapse
Affiliation(s)
- Naoyuki Iida
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Madoka Kawahara
- Omura Satoshi Memorial Institute and Graduate School for Infection Control, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Riku Hirota
- Omura Satoshi Memorial Institute and Graduate School for Infection Control, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Yoshio Shibagaki
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Seisuke Hattori
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Yuko Morikawa
- Omura Satoshi Memorial Institute and Graduate School for Infection Control, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
8
|
Moonmuang S, Maniratanachote R, Chetprayoon P, Sornsuwan K, Thongkum W, Chupradit K, Tayapiwatana C. Specific Interaction of DARPin with HIV-1 CA NTD Disturbs the Distribution of Gag, RNA Packaging, and Tetraspanin Remodelling in the Membrane. Viruses 2022; 14:v14040824. [PMID: 35458554 PMCID: PMC9025900 DOI: 10.3390/v14040824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/22/2022] Open
Abstract
A designed repeat scaffold protein (AnkGAG1D4) recognizing the human immunodeficiency virus-1 (HIV-1) capsid (CA) was formerly established with antiviral assembly. Here, we investigated the molecular mechanism of AnkGAG1D4 function during the late stages of the HIV-1 replication cycle. By applying stimulated emission-depletion (STED) microscopy, Gag polymerisation was interrupted at the plasma membrane. Disturbance of Gag polymerisation triggered Gag accumulation inside producer cells and trapping of the CD81 tetraspanin on the plasma membrane. Moreover, reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) experiments were performed to validate the packaging efficiency of RNAs. Our results advocated that AnkGAG1D4 interfered with the Gag precursor protein from selecting HIV-1 and cellular RNAs for encapsidation into viral particles. These findings convey additional information on the antiviral activity of AnkGAG1D4 at late stages of the HIV-1 life cycle, which is potential for an alternative anti-HIV molecule.
Collapse
Affiliation(s)
- Sutpirat Moonmuang
- Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (S.M.); (K.S.); (W.T.); (K.C.)
- Department of Medical Technology, Division of Clinical Immunology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Rawiwan Maniratanachote
- Toxicology and Bio Evaluation Service Center (TBES), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (R.M.); (P.C.)
| | - Paninee Chetprayoon
- Toxicology and Bio Evaluation Service Center (TBES), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (R.M.); (P.C.)
| | - Kanokporn Sornsuwan
- Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (S.M.); (K.S.); (W.T.); (K.C.)
| | - Weeraya Thongkum
- Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (S.M.); (K.S.); (W.T.); (K.C.)
- Center of Innovative Immunodiagnostic Development, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Koollawat Chupradit
- Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (S.M.); (K.S.); (W.T.); (K.C.)
- Siriraj Center for Regenerative Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chatchai Tayapiwatana
- Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (S.M.); (K.S.); (W.T.); (K.C.)
- Department of Medical Technology, Division of Clinical Immunology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Innovative Immunodiagnostic Development, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence:
| |
Collapse
|
9
|
Sumner C, Ono A. Relationship between HIV-1 Gag Multimerization and Membrane Binding. Viruses 2022; 14:v14030622. [PMID: 35337029 PMCID: PMC8949992 DOI: 10.3390/v14030622] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/11/2022] Open
Abstract
HIV-1 viral particle assembly occurs specifically at the plasma membrane and is driven primarily by the viral polyprotein Gag. Selective association of Gag with the plasma membrane is a key step in the viral assembly pathway, which is traditionally attributed to the MA domain. MA regulates specific plasma membrane binding through two primary mechanisms including: (1) specific interaction of the MA highly basic region (HBR) with the plasma membrane phospholipid phosphatidylinositol (4,5) bisphosphate [PI(4,5)P2], and (2) tRNA binding to the MA HBR, which prevents Gag association with non-PI(4,5)P2 containing membranes. Gag multimerization, driven by both CA–CA inter-protein interactions and NC-RNA binding, also plays an essential role in viral particle assembly, mediating the establishment and growth of the immature Gag lattice on the plasma membrane. In addition to these functions, the multimerization of HIV-1 Gag has also been demonstrated to enhance its membrane binding activity through the MA domain. This review provides an overview of the mechanisms regulating Gag membrane binding through the MA domain and multimerization through the CA and NC domains, and examines how these two functions are intertwined, allowing for multimerization mediated enhancement of Gag membrane binding.
Collapse
|
10
|
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
|
11
|
Cholesterol plays a decisive role in tetraspanin assemblies during bilayer deformations. Biosystems 2021; 209:104505. [PMID: 34403719 DOI: 10.1016/j.biosystems.2021.104505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/30/2021] [Accepted: 08/07/2021] [Indexed: 11/22/2022]
Abstract
The tetraspanin family plays key roles in many physiological processes, such as, tumour invasion, cell motility, virus infection, cell attachment and entry. Tetraspanins function as molecular scaffolds organized in microdomains with interesting downstream cellular consequences. However, despite their relevance in human physiology, the precise mechanisms of their various functions remain elusive. In particular, the full-length CD81 tetraspanin has interesting cholesterol-related properties that modulate its activity in cells. In this work, we study the opening transition of CD81 under different conditions. We propose that such conformational change is a collaborative process enhanced by simultaneous interactions between multiple identical CD81 tetraspanins. With molecular dynamics simulations we describe the crucial role of a ternary lipid bilayer with cholesterol in CD81 conformational dynamics, observing two emergent properties: first, clusters of CD81 collectively segregate one tetraspanin while favouring one opening transition, second, cumulative cholesterol sequestering by CD81 tetraspanins inhibits large membrane deformations due to local density variations.
Collapse
|
12
|
CD82 and Gangliosides Tune CD81 Membrane Behavior. Int J Mol Sci 2021; 22:ijms22168459. [PMID: 34445169 PMCID: PMC8395132 DOI: 10.3390/ijms22168459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
Tetraspanins are a family of transmembrane proteins that form a network of protein–protein interactions within the plasma membrane. Within this network, tetraspanin are thought to control the lateral segregation of their partners at the plasma membrane through mechanisms involving specific lipids. Here, we used a single molecule tracking approach to study the membrane behavior of tetraspanins in mammary epithelial cells and demonstrate that despite a common overall behavior, each tetraspanin (CD9, CD81 and CD82) has a specific signature in terms of dynamics. Furthermore, we demonstrated that tetraspanin dynamics on the cell surface are dependent on gangliosides. More specifically, we found that CD82 expression increases the dynamics of CD81 and alters its localization at the plasma membrane, this has no effect on the behavior of CD9. Our results provide new information on the ability of CD82 and gangliosides to differentially modulate the dynamics and organization of tetraspanins at the plasma membrane and highlight that its lipid and protein composition is involved in the dynamical architecture of the tetraspanin web. We predict that CD82 may act as a regulator of the lateral segregation of specific tetraspanins at the plasma membrane while gangliosides could play a crucial role in establishing tetraspanin-enriched areas.
Collapse
|
13
|
Amiar S, Husby ML, Wijesinghe KJ, Angel S, Bhattarai N, Gerstman BS, Chapagain PP, Li S, Stahelin RV. Lipid-specific oligomerization of the Marburg virus matrix protein VP40 is regulated by two distinct interfaces for virion assembly. J Biol Chem 2021; 296:100796. [PMID: 34019871 PMCID: PMC8191294 DOI: 10.1016/j.jbc.2021.100796] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 02/03/2023] Open
Abstract
Marburg virus (MARV) is a lipid-enveloped virus harboring a negative-sense RNA genome, which has caused sporadic outbreaks of viral hemorrhagic fever in sub-Saharan Africa. MARV assembles and buds from the host cell plasma membrane where MARV matrix protein (mVP40) dimers associate with anionic lipids at the plasma membrane inner leaflet and undergo a dynamic and extensive self-oligomerization into the structural matrix layer. The MARV matrix layer confers the virion filamentous shape and stability but how host lipids modulate mVP40 oligomerization is mostly unknown. Using in vitro and cellular techniques, we present a mVP40 assembly model highlighting two distinct oligomerization interfaces: the (N-terminal domain [NTD] and C-terminal domain [CTD]) in mVP40. Cellular studies of NTD and CTD oligomerization interface mutants demonstrate the importance of each interface in matrix assembly. The assembly steps include protein trafficking to the plasma membrane, homo-multimerization that induced protein enrichment, plasma membrane fluidity changes, and elongations at the plasma membrane. An ascorbate peroxidase derivative (APEX)-transmission electron microscopy method was employed to closely assess the ultrastructural localization and formation of viral particles for wildtype mVP40 and NTD and CTD oligomerization interface mutants. Taken together, these studies present a mechanistic model of mVP40 oligomerization and assembly at the plasma membrane during virion assembly that requires interactions with phosphatidylserine for NTD–NTD interactions and phosphatidylinositol-4,5-bisphosphate for proper CTD–CTD interactions. These findings have broader implications in understanding budding of lipid-enveloped viruses from the host cell plasma membrane and potential strategies to target protein–protein or lipid–protein interactions to inhibit virus budding.
Collapse
Affiliation(s)
- Souad Amiar
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - Monica L Husby
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - Kaveesha J Wijesinghe
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Stephanie Angel
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - Nisha Bhattarai
- Department of Physics, Florida International University, Miami, Florida, USA
| | - Bernard S Gerstman
- Department of Physics, Florida International University, Miami, Florida, USA; Biomolecular Sciences Institute, Florida International University, Miami, Florida, USA
| | - Prem P Chapagain
- Department of Physics, Florida International University, Miami, Florida, USA; Biomolecular Sciences Institute, Florida International University, Miami, Florida, USA
| | - Sheng Li
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Robert V Stahelin
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
14
|
Rendezvous at Plasma Membrane: Cellular Lipids and tRNA Set up Sites of HIV-1 Particle Assembly and Incorporation of Host Transmembrane Proteins. Viruses 2020; 12:v12080842. [PMID: 32752131 PMCID: PMC7472227 DOI: 10.3390/v12080842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/28/2022] Open
Abstract
The HIV-1 structural polyprotein Gag drives the virus particle assembly specifically at the plasma membrane (PM). During this process, the nascent virion incorporates specific subsets of cellular lipids and host membrane proteins, in addition to viral glycoproteins and viral genomic RNA. Gag binding to the PM is regulated by cellular factors, including PM-specific phospholipid PI(4,5)P2 and tRNAs, both of which bind the highly basic region in the matrix domain of Gag. In this article, we review our current understanding of the roles played by cellular lipids and tRNAs in specific localization of HIV-1 Gag to the PM. Furthermore, we examine the effects of PM-bound Gag on the organization of the PM bilayer and discuss how the reorganization of the PM at the virus assembly site potentially contributes to the enrichment of host transmembrane proteins in the HIV-1 particle. Since some of these host transmembrane proteins alter release, attachment, or infectivity of the nascent virions, the mechanism of Gag targeting to the PM and the nature of virus assembly sites have major implications in virus spread.
Collapse
|
15
|
Sengupta P, Lippincott-Schwartz J. Revisiting Membrane Microdomains and Phase Separation: A Viral Perspective. Viruses 2020; 12:v12070745. [PMID: 32664429 PMCID: PMC7412473 DOI: 10.3390/v12070745] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
Retroviruses selectively incorporate a specific subset of host cell proteins and lipids into their outer membrane when they bud out from the host plasma membrane. This specialized viral membrane composition is critical for both viral survivability and infectivity. Here, we review recent findings from live cell imaging of single virus assembly demonstrating that proteins and lipids sort into the HIV retroviral membrane by a mechanism of lipid-based phase partitioning. The findings showed that multimerizing HIV Gag at the assembly site creates a liquid-ordered lipid phase enriched in cholesterol and sphingolipids. Proteins with affinity for this specialized lipid environment partition into it, resulting in the selective incorporation of proteins into the nascent viral membrane. Building on this and other work in the field, we propose a model describing how HIV Gag induces phase separation of the viral assembly site through a mechanism involving transbilayer coupling of lipid acyl chains and membrane curvature changes. Similar phase-partitioning pathways in response to multimerizing structural proteins likely help sort proteins into the membranes of other budding structures within cells.
Collapse
|
16
|
Okutomi T, Minakawa S, Hirota R, Katagiri K, Morikawa Y. HIV Reactivation in Latently Infected Cells With Virological Synapse-Like Cell Contact. Viruses 2020; 12:v12040417. [PMID: 32276457 PMCID: PMC7232209 DOI: 10.3390/v12040417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
HIV reactivation from latency is induced by cytokines but also by cell contact with other cells. To better understand this, J1.1 cells, a latent HIV-1-infected Jurkat derivative, were cocultured with its parental Jurkat. J1.1 cells became p17MA-positive and produced a high level of HIV p24CA antigen, only when they were cocultured with stimulated Jurkat with cell-to-cell contact. In contrast, very little p24CA was produced when they were cocultured without cell contact. Similar results were obtained when latent ACH-2 and its parental A3.01 cells were cocultured. Confocal microscopy revealed that not only HIV-1 p17MA and gp120Env but also LFA-1, CD81, CD59, and TCR CD3 accumulated at the cell contact site, suggesting formation of the virological synapse-like structure. LFA-1–ICAM-1 interaction was involved in the cell-to-cell contact. When J1.1 was cocultured with TCR-deficient Jurkat, the p17MA-positive rate was significantly lower, although the cell-to-cell contact was not impaired. Quantitative proteomics identified 54 membrane molecules, one of which was MHC class I, that accumulated at the cell contact site. Reactivation from latency was also influenced by the presence of stromal cells. Our study indicated that latent HIV-1 in J1.1/ACH-2 cells was efficiently reactivated by cell-to-cell contact with stimulated parental cells, accompanying the virological synapse-like structure.
Collapse
Affiliation(s)
- Toshiki Okutomi
- Graduate School of Infection Control Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan; (T.O.); (S.M.); (R.H.)
| | - Satoko Minakawa
- Graduate School of Infection Control Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan; (T.O.); (S.M.); (R.H.)
| | - Riku Hirota
- Graduate School of Infection Control Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan; (T.O.); (S.M.); (R.H.)
| | - Koko Katagiri
- Department of Biosciences, School of Science, Kitasato University, Kitasato 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan;
| | - Yuko Morikawa
- Graduate School of Infection Control Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan; (T.O.); (S.M.); (R.H.)
- Correspondence: ; Tel.: +81-3-5791-6129
| |
Collapse
|
17
|
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
|
18
|
Chavent M, Duncan AL, Rassam P, Birkholz O, Hélie J, Reddy T, Beliaev D, Hambly B, Piehler J, Kleanthous C, Sansom MSP. How nanoscale protein interactions determine the mesoscale dynamic organisation of bacterial outer membrane proteins. Nat Commun 2018; 9:2846. [PMID: 30030429 PMCID: PMC6054660 DOI: 10.1038/s41467-018-05255-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 06/15/2018] [Indexed: 01/07/2023] Open
Abstract
The spatiotemporal organisation of membranes is often characterised by the formation of large protein clusters. In Escherichia coli, outer membrane protein (OMP) clustering leads to OMP islands, the formation of which underpins OMP turnover and drives organisation across the cell envelope. Modelling how OMP islands form in order to understand their origin and outer membrane behaviour has been confounded by the inherent difficulties of simulating large numbers of OMPs over meaningful timescales. Here, we overcome these problems by training a mesoscale model incorporating thousands of OMPs on coarse-grained molecular dynamics simulations. We achieve simulations over timescales that allow direct comparison to experimental data of OMP behaviour. We show that specific interaction surfaces between OMPs are key to the formation of OMP clusters, that OMP clusters present a mesh of moving barriers that confine newly inserted proteins within islands, and that mesoscale simulations recapitulate the restricted diffusion characteristics of OMPs. In Escherichia coli, outer membrane protein (OMP) cluster and form islands, but the origin and behaviour of those clusters remains poorly understood. Here authors use coarse grained molecular dynamics simulation and show that their mesoscale simulations recapitulate the restricted diffusion characteristics of OMPs.
Collapse
Affiliation(s)
- Matthieu Chavent
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK.,Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 205 Route de Narbonne, Toulouse, 31400, France
| | - Anna L Duncan
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK
| | - Patrice Rassam
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK.,Laboratoire de Biophotonique et Pharmacologie, UMR 7213 CNRS, Faculté de Pharmacie, Université de Strasbourg, 74 Route du Rhin, 67401, Illkirch, France
| | - Oliver Birkholz
- Department of Biology, University of Osnabrück, Barbarastraße 11, 49076, Osnabrück, Germany
| | - Jean Hélie
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK.,SEMMLE, Blue Boar Court, 9 Alfred St, Oxford, OX1 4EH, UK
| | - Tyler Reddy
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK.,Theoretical Biology and Biophysics, T-6, Los Alamos National Laboratory, Los Alamos, NM, 87525, USA
| | - Dmitry Beliaev
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter (550), Woodstock Road, Oxford, OX2 6GG, UK
| | - Ben Hambly
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter (550), Woodstock Road, Oxford, OX2 6GG, UK
| | - Jacob Piehler
- Department of Biology, University of Osnabrück, Barbarastraße 11, 49076, Osnabrück, Germany
| | - Colin Kleanthous
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK.
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK.
| |
Collapse
|
19
|
Thomson-Luque R, Wang C, Ntumngia FB, Xu S, Szekeres K, Conway A, Adapa SR, Barnes SJ, Adams JH, Jiang RHY. In-depth phenotypic characterization of reticulocyte maturation using mass cytometry. Blood Cells Mol Dis 2018; 72:22-33. [PMID: 30007855 PMCID: PMC6097872 DOI: 10.1016/j.bcmd.2018.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/24/2018] [Accepted: 06/24/2018] [Indexed: 12/17/2022]
Abstract
Progress towards an in-depth understanding of the final steps of the erythroid lineage development is paramount for many hematological diseases. We have characterized the final stages of reticulocyte maturation from bone marrow to peripheral blood using for the first time single-cell Mass Cytometry (CyTOF). We were able to measure the expression of 31 surface markers within a single red blood cell (RBC). We demonstrate the validity of CyTOF for RBC phenotyping by confirming the progressive reduction of transferrin receptor 1 (CD71) during reticulocyte maturation to mature RBC. We highlight the high-dimensional nature of mass cytometry data by correlating the expression of multiple proteins on individual RBCs. We further describe a more drastic reduction pattern for a component of the alpha4/beta1 integrin CD49d at the very early steps of reticulocyte maturation in bone marrow and directly linked with the mitochondria remnants clearance pattern. The enhanced and accurate RBC phenotyping potential of CyTOF described herein could be beneficial to decipher RBC preferences, as well as still not well understood receptor-ligand interaction of some hemotropic parasites such as the malaria causing agent Plasmodium vivax.
Collapse
Affiliation(s)
- Richard Thomson-Luque
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Chengqi Wang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Francis B Ntumngia
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Shulin Xu
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Karoly Szekeres
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Amy Conway
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Swamy Rakesh Adapa
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - Samantha J Barnes
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA.
| | - Rays H Y Jiang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, USA.
| |
Collapse
|
20
|
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: 80] [Impact Index Per Article: 11.4] [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
|
21
|
Doktorova M, Heberle FA, Kingston RL, Khelashvili G, Cuendet MA, Wen Y, Katsaras J, Feigenson GW, Vogt VM, Dick RA. Cholesterol Promotes Protein Binding by Affecting Membrane Electrostatics and Solvation Properties. Biophys J 2017; 113:2004-2015. [PMID: 29117524 DOI: 10.1016/j.bpj.2017.08.055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 01/28/2023] Open
Abstract
Binding of the retroviral structural protein Gag to the cellular plasma membrane is mediated by the protein's matrix (MA) domain. Prominent among MA-PM interactions is electrostatic attraction between the positively charged MA domain and the negatively charged plasma membrane inner leaflet. Previously, we reported that membrane association of HIV-1 Gag, as well as purified Rous sarcoma virus (RSV) MA and Gag, depends strongly on the presence of acidic lipids and is enhanced by cholesterol (Chol). The mechanism underlying this enhancement was unclear. Here, using a broad set of in vitro and in silico techniques we addressed molecular mechanisms of association between RSV MA and model membranes, and investigated how Chol enhances this association. In neutron scattering experiments with liposomes in the presence or absence of Chol, MA preferentially interacted with preexisting POPS-rich clusters formed by nonideal lipid mixing, binding peripherally to the lipid headgroups with minimal perturbation to the bilayer structure. Molecular dynamics simulations showed a stronger MA-bilayer interaction in the presence of Chol, and a large Chol-driven increase in lipid packing and membrane surface charge density. Although in vitro MA-liposome association is influenced by disparate variables, including ionic strength and concentrations of Chol and charged lipids, continuum electrostatic theory revealed an underlying dependence on membrane surface potential. Together, these results conclusively show that Chol affects RSV MA-membrane association by making the electrostatic potential at the membrane surface more negative, while decreasing the penalty for lipid headgroup desolvation. The presented approach can be applied to other viral and nonviral proteins.
Collapse
Affiliation(s)
- Milka Doktorova
- Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York
| | - Frederick A Heberle
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee; Joint Institute for Biological Sciences, Oak Ridge National Laboratory, Oak Ridge, Tennessee; Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee.
| | - Richard L Kingston
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Michel A Cuendet
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Yi Wen
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York
| | - John Katsaras
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee; Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee; Department of Physics and Astronomy, University of Tennessee, Knoxville, Tennessee; Shull Wollan Center, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Gerald W Feigenson
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York
| | - Volker M Vogt
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York
| | - Robert A Dick
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York.
| |
Collapse
|
22
|
Schneider IC, Hartmann J, Braun G, Stitz J, Klamp T, Bihi M, Sahin U, Buchholz CJ. Displaying Tetra-Membrane Spanning Claudins on Enveloped Virus-Like Particles for Cancer Immunotherapy. Biotechnol J 2017; 13:e1700345. [DOI: 10.1002/biot.201700345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 11/06/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Irene C. Schneider
- Molecular Biotechnology and Gene Therapy; Paul-Ehrlich-Institut; Langen Germany
| | - Jessica Hartmann
- Molecular Biotechnology and Gene Therapy; Paul-Ehrlich-Institut; Langen Germany
| | - Gundula Braun
- Molecular Biotechnology and Gene Therapy; Paul-Ehrlich-Institut; Langen Germany
| | - Jörn Stitz
- Faculty of Applied Natural Sciences, TH Köln; University of Applied Sciences Cologne; Leverkusen Germany
| | | | - Mahjoub Bihi
- BioNTech Protein Therapeutics GmbH; Mainz Germany
| | - Ugur Sahin
- BioNTech Protein Therapeutics GmbH; Mainz Germany
| | | |
Collapse
|
23
|
Doktorova M, Heberle FA, Kingston RL, Khelashvili G, Cuendet MA, Wen Y, Katsaras J, Feigenson GW, Vogt VM, Dick RA. Cholesterol Promotes Protein Binding by Affecting Membrane Electrostatics and Solvation Properties. Biophys J 2017. [PMID: 29117524 DOI: 10.1016/j.bpj.2017.08.055.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Binding of the retroviral structural protein Gag to the cellular plasma membrane is mediated by the protein's matrix (MA) domain. Prominent among MA-PM interactions is electrostatic attraction between the positively charged MA domain and the negatively charged plasma membrane inner leaflet. Previously, we reported that membrane association of HIV-1 Gag, as well as purified Rous sarcoma virus (RSV) MA and Gag, depends strongly on the presence of acidic lipids and is enhanced by cholesterol (Chol). The mechanism underlying this enhancement was unclear. Here, using a broad set of in vitro and in silico techniques we addressed molecular mechanisms of association between RSV MA and model membranes, and investigated how Chol enhances this association. In neutron scattering experiments with liposomes in the presence or absence of Chol, MA preferentially interacted with preexisting POPS-rich clusters formed by nonideal lipid mixing, binding peripherally to the lipid headgroups with minimal perturbation to the bilayer structure. Molecular dynamics simulations showed a stronger MA-bilayer interaction in the presence of Chol, and a large Chol-driven increase in lipid packing and membrane surface charge density. Although in vitro MA-liposome association is influenced by disparate variables, including ionic strength and concentrations of Chol and charged lipids, continuum electrostatic theory revealed an underlying dependence on membrane surface potential. Together, these results conclusively show that Chol affects RSV MA-membrane association by making the electrostatic potential at the membrane surface more negative, while decreasing the penalty for lipid headgroup desolvation. The presented approach can be applied to other viral and nonviral proteins.
Collapse
Affiliation(s)
- Milka Doktorova
- Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York
| | - Frederick A Heberle
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee; Joint Institute for Biological Sciences, Oak Ridge National Laboratory, Oak Ridge, Tennessee; Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee.
| | - Richard L Kingston
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Michel A Cuendet
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Yi Wen
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York
| | - John Katsaras
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee; Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee; Department of Physics and Astronomy, University of Tennessee, Knoxville, Tennessee; Shull Wollan Center, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Gerald W Feigenson
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York
| | - Volker M Vogt
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York
| | - Robert A Dick
- Department of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca, New York.
| |
Collapse
|
24
|
Haining EJ, Matthews AL, Noy PJ, Romanska HM, Harris HJ, Pike J, Morowski M, Gavin RL, Yang J, Milhiet PE, Berditchevski F, Nieswandt B, Poulter NS, Watson SP, Tomlinson MG. Tetraspanin Tspan9 regulates platelet collagen receptor GPVI lateral diffusion and activation. Platelets 2017; 28:629-642. [PMID: 28032533 PMCID: PMC5706974 DOI: 10.1080/09537104.2016.1254175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/06/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022]
Abstract
The tetraspanins are a superfamily of four-transmembrane proteins, which regulate the trafficking, lateral diffusion and clustering of the transmembrane proteins with which they interact. We have previously shown that tetraspanin Tspan9 is expressed on platelets. Here we have characterised gene-trap mice lacking Tspan9. The mice were viable with normal platelet numbers and size. Tspan9-deficient platelets were specifically defective in aggregation and secretion induced by the platelet collagen receptor GPVI, despite normal surface GPVI expression levels. A GPVI activation defect was suggested by partially impaired GPVI-induced protein tyrosine phosphorylation. In mechanistic experiments, Tspan9 and GPVI co-immunoprecipitated and co-localised, but super-resolution imaging revealed no defects in collagen-induced GPVI clustering on Tspan9-deficient platelets. However, single particle tracking using total internal reflection fluorescence microscopy showed that GPVI lateral diffusion was reduced by approximately 50% in the absence of Tspan9. Therefore, Tspan9 plays a fine-tuning role in platelet activation by regulating GPVI membrane dynamics.
Collapse
Affiliation(s)
- Elizabeth J. Haining
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Alexandra L. Matthews
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Peter J. Noy
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | | | - Helen J. Harris
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Jeremy Pike
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
- PSIBS Doctoral Training Centre, School of Chemistry, University of Birmingham, Birmingham, UK
| | - Martina Morowski
- Department of Experimental Biomedicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Rebecca L. Gavin
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Jing Yang
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Pierre-Emmanuel Milhiet
- INSERM U1054, CNRS, UMR 5048, Centre de Biochimie Structurale, Montpellier University, France
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Bernhard Nieswandt
- Department of Experimental Biomedicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Natalie S. Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Michael G. Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
25
|
Earnest JT, Hantak MP, Li K, McCray PB, Perlman S, Gallagher T. The tetraspanin CD9 facilitates MERS-coronavirus entry by scaffolding host cell receptors and proteases. PLoS Pathog 2017; 13:e1006546. [PMID: 28759649 PMCID: PMC5552337 DOI: 10.1371/journal.ppat.1006546] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/10/2017] [Accepted: 07/21/2017] [Indexed: 01/27/2023] Open
Abstract
Infection by enveloped coronaviruses (CoVs) initiates with viral spike (S) proteins binding to cellular receptors, and is followed by proteolytic cleavage of receptor-bound S proteins, which prompts S protein-mediated virus-cell membrane fusion. Infection therefore requires close proximity of receptors and proteases. We considered whether tetraspanins, scaffolding proteins known to facilitate CoV infections, hold receptors and proteases together on cell membranes. Using knockout cell lines, we found that the tetraspanin CD9, but not the tetraspanin CD81, formed cell-surface complexes of dipeptidyl peptidase 4 (DPP4), the MERS-CoV receptor, and the type II transmembrane serine protease (TTSP) member TMPRSS2, a CoV-activating protease. This CD9-facilitated condensation of receptors and proteases allowed MERS-CoV pseudoviruses to enter cells rapidly and efficiently. Without CD9, MERS-CoV viruses were not activated by TTSPs, and they trafficked into endosomes to be cleaved much later and less efficiently by cathepsins. Thus, we identified DPP4:CD9:TTSP as the protein complexes necessary for early, efficient MERS-CoV entry. To evaluate the importance of these complexes in an in vivo CoV infection model, we used recombinant Adenovirus 5 (rAd5) vectors to express human DPP4 in mouse lungs, thereby sensitizing the animals to MERS-CoV infection. When the rAd5-hDPP4 vectors co-expressed small RNAs silencing Cd9 or Tmprss2, the animals were significantly less susceptible, indicating that CD9 and TMPRSS2 facilitated robust in vivo MERS-CoV infection of mouse lungs. Furthermore, the S proteins of virulent mouse-adapted MERS-CoVs acquired a CD9-dependent cell entry character, suggesting that CD9 is a selective agent in the evolution of CoV virulence.
Collapse
Affiliation(s)
- James T. Earnest
- Department of Microbiology and Immunology, Loyola University Medical Center, Maywood, IL, United States of America
| | - Michael P. Hantak
- Department of Microbiology and Immunology, Loyola University Medical Center, Maywood, IL, United States of America
| | - Kun Li
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Paul B. McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Stanley Perlman
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Department of Microbiology, University of Iowa, Iowa City, IA, United States of America
| | - Tom Gallagher
- Department of Microbiology and Immunology, Loyola University Medical Center, Maywood, IL, United States of America
- * E-mail:
| |
Collapse
|
26
|
Inlora J, Chukkapalli V, Bedi S, Ono A. Molecular Determinants Directing HIV-1 Gag Assembly to Virus-Containing Compartments in Primary Macrophages. J Virol 2016; 90:8509-19. [PMID: 27440886 PMCID: PMC5021390 DOI: 10.1128/jvi.01004-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/11/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED The subcellular sites of HIV-1 assembly, determined by the localization of the structural protein Gag, vary in a cell-type-dependent manner. In T cells and transformed cell lines used as model systems, HIV-1 assembles at the plasma membrane (PM). The binding and localization of HIV-1 Gag to the PM are mediated by the interaction between the matrix (MA) domain, specifically the highly basic region, and a PM-specific acidic phospholipid, phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2]. In primary macrophages, prominent accumulation of assembling or assembled particles is found in the virus-containing compartments (VCCs), which largely consist of convoluted invaginations of the PM. To elucidate the molecular mechanism of HIV-1 Gag targeting to the VCCs, we examined the impact of overexpression of polyphosphoinositide 5-phosphatase IV (5ptaseIV), which depletes cellular PI(4,5)P2, in primary macrophages. We found that the VCC localization and virus release of HIV-1 are severely impaired upon 5ptaseIV overexpression, suggesting an important role for the MA-PI(4,5)P2 interaction in HIV-1 assembly in primary macrophages. However, our analysis of HIV-1 Gag derivatives with MA changes showed that this interaction contributes to Gag membrane binding but is dispensable for specific targeting of Gag to the VCCs per se We further determined that deletion of the NC domain abolishes VCC-specific localization of HIV-1 Gag. Notably, HIV-1 Gag localized efficiently to the VCCs when the NC domain was replaced with a leucine zipper dimerization motif that promotes Gag multimerization. Altogether, our data revealed that targeting of HIV-1 Gag to the VCCs requires NC-dependent multimerization. IMPORTANCE In T cells and model cell lines, HIV-1 Gag localizes to the PM in a manner dependent on the MA-PI(4,5)P2 interaction. On the other hand, in primary macrophages, HIV-1 Gag localizes to convoluted intracellular membrane structures termed virus-containing compartments (VCCs). Although these compartments have been known for decades, and despite the implication of viruses in VCCs being involved in virus reservoir maintenance and spread, the viral determinant(s) that promotes Gag targeting to VCCs is unknown. In this study, we found that the MA-PI(4,5)P2 interaction facilitates efficient Gag membrane binding in macrophages but is not essential for Gag targeting to VCCs. Rather, our results revealed that NC-dependent multimerization promotes VCC targeting. Our findings highlight the differential roles played by MA and NC in HIV-1 Gag membrane binding and targeting and suggest a multimerization-dependent mechanism for Gag trafficking in primary macrophages similar to that for Gag localization to uropods in polarized T cells.
Collapse
Affiliation(s)
- Jingga Inlora
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Vineela Chukkapalli
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sukhmani Bedi
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
27
|
Matheson NJ, Greenwood EJ, Lehner PJ. Manipulation of immunometabolism by HIV-accessories to the crime? Curr Opin Virol 2016; 19:65-70. [PMID: 27448768 DOI: 10.1016/j.coviro.2016.06.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022]
Abstract
Evolutionary pressure has produced an 'arms race' between cellular restriction factors (limiting viral replication) and viral proteins (overcoming host restriction). The host factors SAMHD1 and SLFN1 patrol metabolic bottlenecks required for HIV replication. Conversely, the HIV accessory proteins Vpx, Vpu and Nef manipulate cellular metabolism to enable viral replication. Recent work identifying Vpu-mediated downregulation of the alanine transporter SNAT1 and Nef-mediated downregulation of the serine carriers SERINC3/5 has uncovered the importance of HIV manipulation of the amino acid supply. Interference with CD4(+) T-cell amino acid metabolism suggests a novel paradigm of viral immunomodulation, and signposts fundamental aspects of lymphocyte biology.
Collapse
Affiliation(s)
- Nicholas J Matheson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK.
| | - Edward Jd Greenwood
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Paul J Lehner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK.
| |
Collapse
|
28
|
Dosset P, Rassam P, Fernandez L, Espenel C, Rubinstein E, Margeat E, Milhiet PE. Automatic detection of diffusion modes within biological membranes using back-propagation neural network. BMC Bioinformatics 2016; 17:197. [PMID: 27141816 PMCID: PMC4855490 DOI: 10.1186/s12859-016-1064-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 04/29/2016] [Indexed: 12/17/2022] Open
Abstract
Background Single particle tracking (SPT) is nowadays one of the most popular technique to probe spatio-temporal dynamics of proteins diffusing within the plasma membrane. Indeed membrane components of eukaryotic cells are very dynamic molecules and can diffuse according to different motion modes. Trajectories are often reconstructed frame-by-frame and dynamic properties often evaluated using mean square displacement (MSD) analysis. However, to get statistically significant results in tracking experiments, analysis of a large number of trajectories is required and new methods facilitating this analysis are still needed. Results In this study we developed a new algorithm based on back-propagation neural network (BPNN) and MSD analysis using a sliding window. The neural network was trained and cross validated with short synthetic trajectories. For simulated and experimental data, the algorithm was shown to accurately discriminate between Brownian, confined and directed diffusion modes within one trajectory, the 3 main of diffusion encountered for proteins diffusing within biological membranes. It does not require a minimum number of observed particle displacements within the trajectory to infer the presence of multiple motion states. The size of the sliding window was small enough to measure local behavior and to detect switches between different diffusion modes for segments as short as 20 frames. It also provides quantitative information from each segment of these trajectories. Besides its ability to detect switches between 3 modes of diffusion, this algorithm is able to analyze simultaneously hundreds of trajectories with a short computational time. Conclusion This new algorithm, implemented in powerful and handy software, provides a new conceptual and versatile tool, to accurately analyze the dynamic behavior of membrane components. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1064-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patrice Dosset
- Inserm, U1054, Montpellier, France.,Université de Montpellier, CNRS, UMR 5048, Centre de Biochimie Structurale, Montpellier, France
| | - Patrice Rassam
- Inserm, U1054, Montpellier, France.,Université de Montpellier, CNRS, UMR 5048, Centre de Biochimie Structurale, Montpellier, France
| | - Laurent Fernandez
- Inserm, U1054, Montpellier, France.,Université de Montpellier, CNRS, UMR 5048, Centre de Biochimie Structurale, Montpellier, France
| | - Cedric Espenel
- Department of Cell and Developmental Biology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Eric Rubinstein
- Inserm, U1004, Villejuif, France.,Institut André Lwoff, Université Paris 11, Villejuif, France
| | - Emmanuel Margeat
- Inserm, U1054, Montpellier, France.,Université de Montpellier, CNRS, UMR 5048, Centre de Biochimie Structurale, Montpellier, France
| | - Pierre-Emmanuel Milhiet
- Inserm, U1054, Montpellier, France. .,Université de Montpellier, CNRS, UMR 5048, Centre de Biochimie Structurale, Montpellier, France. .,Centre de Biochimie Structurale, 29, rue de Navacelles, 34090, Montpellier, France.
| |
Collapse
|
29
|
Chang GW, Hsiao CC, Peng YM, Vieira Braga F, Kragten N, Remmerswaal E, van de Garde M, Straussberg R, König G, Kostenis E, Knäuper V, Meyaard L, van Lier R, van Gisbergen K, Lin HH, Hamann J. The Adhesion G Protein-Coupled Receptor GPR56/ADGRG1 Is an Inhibitory Receptor on Human NK Cells. Cell Rep 2016; 15:1757-70. [DOI: 10.1016/j.celrep.2016.04.053] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 01/22/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022] Open
|
30
|
Florian PE, Rouillé Y, Ruta S, Nichita N, Roseanu A. Recent advances in human viruses imaging studies. J Basic Microbiol 2016; 56:591-607. [PMID: 27059598 DOI: 10.1002/jobm.201500575] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/27/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Paula Ecaterina Florian
- Department of ; Ligand-Receptor Interactions; Institute of Biochemistry of the Romanian Academy; Bucharest Romania
| | - Yves Rouillé
- Center for Infection and Immunity of Lille (CIIL); Inserm U1019; CNRS UMR8204; Institut Pasteur de Lille; Université Lille Nord de France; Lille France
| | - Simona Ruta
- Department of Emergent Diseases; Stefan S. Nicolau Institute of Virology; Bucharest 030304 Romania
| | - Norica Nichita
- Department of Viral Glycoproteins; Institute of Biochemistry of the Romanian Academy; Bucharest Romania
| | - Anca Roseanu
- Department of ; Ligand-Receptor Interactions; Institute of Biochemistry of the Romanian Academy; Bucharest Romania
| |
Collapse
|
31
|
Soares HR, Castro R, Tomás HA, Rodrigues AF, Gomes-Alves P, Bellier B, Klatzmann D, Carrondo MJT, Alves PM, Coroadinha AS. Tetraspanins displayed in retrovirus-derived virus-like particles and their immunogenicity. Vaccine 2016; 34:1634-1641. [PMID: 26795367 DOI: 10.1016/j.vaccine.2015.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 10/22/2022]
Abstract
Virus-like particles (VLPs) are a particular subset of subunit vaccines which are currently explored as safer alternatives to live attenuated or inactivated vaccines. VLPs derived from retrovirus (retroVLPs) are commonly used as scaffolds for vaccine candidates due to their ability to incorporate heterologous envelope proteins. Pseudotyping retroVLPs is however not a selective process therefore, host cellular proteins such as tetraspanins are also included in the membrane. The contribution of these host-proteins to retrovirus immunogenicity remains unclear. In this work, human cells silenced and not silenced for tetraspanin CD81 were used to produce CD81(-) or CD81(+) retroVLPs. We first analyzed mice immune response against human CD81. Despite effective silencing of CD81 in retroVLP producing cells, both humoral and cellular immune responses showed persistent anti-CD81 immunogenicity, suggesting cross reactivity to related antigens. We thus compared the incorporation of related tetraspanins in retroVLPs and showed that decreased CD81 incorporation in CD81(-) retro-VLPs is compensated by an increased incorporation of CD9 and CD63 tetraspanins. These results highlight the dynamic nature of host-derived proteins incorporation in retroVLPs membrane, which should be considered when retrovirus-based biopharmaceuticals are produced in xenogeneic cells.
Collapse
Affiliation(s)
- H R Soares
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - R Castro
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - H A Tomás
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - A F Rodrigues
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - P Gomes-Alves
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - B Bellier
- Sorbonne Universités, UPMC Univ Paris 06, UMRS_959, I3, F-75013 Paris, France; INSERM, UMR_S 959, I3, F-75013 Paris, France
| | - D Klatzmann
- Sorbonne Universités, UPMC Univ Paris 06, UMRS_959, I3, F-75013 Paris, France; INSERM, UMR_S 959, I3, F-75013 Paris, France
| | - M J T Carrondo
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Faculdade de Ciências e Tecnologia/Universidade Nova de Lisboa, P-2825 Monte da Caparica, Portugal
| | - P M Alves
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - A S Coroadinha
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
32
|
Jouannet S, Saint-Pol J, Fernandez L, Nguyen V, Charrin S, Boucheix C, Brou C, Milhiet PE, Rubinstein E. TspanC8 tetraspanins differentially regulate the cleavage of ADAM10 substrates, Notch activation and ADAM10 membrane compartmentalization. Cell Mol Life Sci 2015; 73:1895-915. [PMID: 26686862 PMCID: PMC4819958 DOI: 10.1007/s00018-015-2111-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/03/2015] [Accepted: 12/03/2015] [Indexed: 01/08/2023]
Abstract
The metalloprotease ADAM10 mediates the shedding of the ectodomain of various cell membrane proteins, including APP, the precursor of the amyloid peptide Aβ, and Notch receptors following ligand binding. ADAM10 associates with the members of an evolutionary conserved subgroup of tetraspanins, referred to as TspanC8, which regulate its exit from the endoplasmic reticulum. Here we show that 4 of these TspanC8 (Tspan5, Tspan14, Tspan15 and Tspan33) which positively regulate ADAM10 surface expression levels differentially impact ADAM10-dependent Notch activation and the cleavage of several ADAM10 substrates, including APP, N-cadherin and CD44. Sucrose gradient fractionation, single molecule tracking and quantitative mass-spectrometry analysis of the repertoire of molecules co-immunoprecipitated with Tspan5, Tspan15 and ADAM10 show that these two tetraspanins differentially regulate ADAM10 membrane compartmentalization. These data represent a unique example where several tetraspanins differentially regulate the function of a common partner protein through a distinct membrane compartmentalization.
Collapse
Affiliation(s)
- Stéphanie Jouannet
- Inserm, U935, 94807, Villejuif, France.,Université Paris-Sud, Institut André Lwoff, 94807, Villejuif, France
| | - Julien Saint-Pol
- Inserm, U935, 94807, Villejuif, France.,Université Paris-Sud, Institut André Lwoff, 94807, Villejuif, France
| | - Laurent Fernandez
- Inserm, U1054, 34090, Montpellier, France.,Université de Montpellier, CNRS, UMR5048, Centre de Biochimie Structurale, Montpellier, France
| | - Viet Nguyen
- Université Paris-Sud, Institut André Lwoff, 94807, Villejuif, France
| | - Stéphanie Charrin
- Inserm, U935, 94807, Villejuif, France.,Université Paris-Sud, Institut André Lwoff, 94807, Villejuif, France
| | - Claude Boucheix
- Inserm, U935, 94807, Villejuif, France.,Université Paris-Sud, Institut André Lwoff, 94807, Villejuif, France
| | - Christel Brou
- Institut Pasteur, Laboratoire "Signalisation et Pathogenèse", 75015, Paris, France
| | - Pierre-Emmanuel Milhiet
- Inserm, U1054, 34090, Montpellier, France.,Université de Montpellier, CNRS, UMR5048, Centre de Biochimie Structurale, Montpellier, France
| | - Eric Rubinstein
- Inserm, U935, 94807, Villejuif, France. .,Université Paris-Sud, Institut André Lwoff, 94807, Villejuif, France.
| |
Collapse
|
33
|
Homsi Y, Schloetel JG, Scheffer KD, Schmidt TH, Destainville N, Florin L, Lang T. The extracellular δ-domain is essential for the formation of CD81 tetraspanin webs. Biophys J 2015; 107:100-13. [PMID: 24988345 DOI: 10.1016/j.bpj.2014.05.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 05/14/2014] [Accepted: 05/20/2014] [Indexed: 12/15/2022] Open
Abstract
CD81 is a ubiquitously expressed member of the tetraspanin family. It forms large molecular platforms, so-called tetraspanin webs that play physiological roles in a variety of cellular functions and are involved in viral and parasite infections. We have investigated which part of the CD81 molecule is required for the formation of domains in the cell membranes of T-cells and hepatocytes. Surprisingly, we find that large CD81 platforms assemble via the short extracellular δ-domain, independent from a strong primary partner binding and from weak interactions mediated by palmitoylation. The δ-domain is also essential for the platforms to function during viral entry. We propose that, instead of stable binary interactions, CD81 interactions via the small δ-domain, possibly involving a dimerization step, play the key role in organizing CD81 into large tetraspanin webs and controlling its function.
Collapse
Affiliation(s)
- Yahya Homsi
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jan-Gero Schloetel
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Konstanze D Scheffer
- Department of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas H Schmidt
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Nicolas Destainville
- Université Toulouse 3-Paul Sabatier, UPS, Laboratoire de Physique Théorique (IRSAMC), Toulouse, France
| | - Luise Florin
- Department of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Thorsten Lang
- Department of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
34
|
Rassam P, Copeland NA, Birkholz O, Tóth C, Chavent M, Duncan AL, Cross SJ, Housden NG, Kaminska R, Seger U, Quinn DM, Garrod TJ, Sansom MSP, Piehler J, Baumann CG, Kleanthous C. Supramolecular assemblies underpin turnover of outer membrane proteins in bacteria. Nature 2015; 523:333-6. [PMID: 26061769 PMCID: PMC4905513 DOI: 10.1038/nature14461] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 04/08/2015] [Indexed: 12/24/2022]
Abstract
Gram-negative bacteria inhabit a broad range of ecological niches. For Escherichia coli, this includes river water as well as humans and animals, where it can be both a commensal and a pathogen. Intricate regulatory mechanisms ensure that bacteria have the right complement of β-barrel outer membrane proteins (OMPs) to enable adaptation to a particular habitat. Yet no mechanism is known for replacing OMPs in the outer membrane, an issue that is further confounded by the lack of an energy source and the high stability and abundance of OMPs. Here we uncover the process underpinning OMP turnover in E. coli and show it to be passive and binary in nature, in which old OMPs are displaced to the poles of growing cells as new OMPs take their place. Using fluorescent colicins as OMP-specific probes, in combination with ensemble and single-molecule fluorescence microscopy in vivo and in vitro, as well as molecular dynamics simulations, we established the mechanism for binary OMP partitioning. OMPs clustered to form ∼0.5-μm diameter islands, where their diffusion is restricted by promiscuous interactions with other OMPs. OMP islands were distributed throughout the cell and contained the Bam complex, which catalyses the insertion of OMPs in the outer membrane. However, OMP biogenesis occurred as a gradient that was highest at mid-cell but largely absent at cell poles. The cumulative effect is to push old OMP islands towards the poles of growing cells, leading to a binary distribution when cells divide. Hence, the outer membrane of a Gram-negative bacterium is a spatially and temporally organized structure, and this organization lies at the heart of how OMPs are turned over in the membrane.
Collapse
Affiliation(s)
- Patrice Rassam
- 1] Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK [2] Department of Biology, University of York, York YO10 5DD, UK
| | | | - Oliver Birkholz
- Department of Biology, University of Osnabrück, Barbarastraße 11, 49076 Osnabrück, Germany
| | - Csaba Tóth
- Department of Biology, University of York, York YO10 5DD, UK
| | - Matthieu Chavent
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Anna L Duncan
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Stephen J Cross
- Department of Biology, University of York, York YO10 5DD, UK
| | - Nicholas G Housden
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Renata Kaminska
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Urban Seger
- Department of Biology, University of York, York YO10 5DD, UK
| | - Diana M Quinn
- Department of Biology, University of York, York YO10 5DD, UK
| | - Tamsin J Garrod
- Department of Biology, University of York, York YO10 5DD, UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Jacob Piehler
- Department of Biology, University of Osnabrück, Barbarastraße 11, 49076 Osnabrück, Germany
| | | | - Colin Kleanthous
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| |
Collapse
|
35
|
Colquhoun DR, Lyashkov AE, Ubaida Mohien C, Aquino VN, Bullock BT, Dinglasan RR, Agnew BJ, Graham DRM. Bioorthogonal mimetics of palmitoyl-CoA and myristoyl-CoA and their subsequent isolation by click chemistry and characterization by mass spectrometry reveal novel acylated host-proteins modified by HIV-1 infection. Proteomics 2015; 15:2066-77. [PMID: 25914232 DOI: 10.1002/pmic.201500063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/15/2015] [Accepted: 04/23/2015] [Indexed: 01/28/2023]
Abstract
Protein acylation plays a critical role in protein localization and function. Acylation is essential for human immunodeficiency virus 1 (HIV-1) assembly and budding of HIV-1 from the plasma membrane in lipid raft microdomains and is mediated by myristoylation of the Gag polyprotein and the copackaging of the envelope protein is facilitated by colocalization mediated by palmitoylation. Since the viral accessory protein NEF has been shown to alter the substrate specificity of myristoyl transferases, and alter cargo trafficking lipid rafts, we hypothesized that HIV-1 infection may alter protein acylation globally. To test this hypothesis, we labeled HIV-1 infected cells with biomimetics of acyl azides, which are incorporated in a manner analogous to natural acyl-Co-A. A terminal azide group allowed us to use a copper catalyzed click chemistry to conjugate the incorporated modifications to a number of substrates to carry out SDS-PAGE, fluorescence microscopy, and enrichment for LC-MS/MS. Using LC-MS/MS, we identified 103 and 174 proteins from the myristic and palmitic azide enrichments, with 27 and 45 proteins respectively that differentiated HIV-1 infected from uninfected cells. This approach has provided us with important insights into HIV-1 biology and is widely applicable to many virological systems.
Collapse
Affiliation(s)
- David R Colquhoun
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexey E Lyashkov
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ceereena Ubaida Mohien
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Veronica N Aquino
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brandon T Bullock
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rhoel R Dinglasan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Brian J Agnew
- Biosciences Group, Thermo Fisher Scientific, Eugene, OR, USA
| | - David R M Graham
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Gerber PP, Cabrini M, Jancic C, Paoletti L, Banchio C, von Bilderling C, Sigaut L, Pietrasanta LI, Duette G, Freed EO, Basile GDS, Moita CF, Moita LF, Amigorena S, Benaroch P, Geffner J, Ostrowski M. Rab27a controls HIV-1 assembly by regulating plasma membrane levels of phosphatidylinositol 4,5-bisphosphate. J Cell Biol 2015; 209:435-52. [PMID: 25940347 PMCID: PMC4427790 DOI: 10.1083/jcb.201409082] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 04/01/2015] [Indexed: 12/22/2022] Open
Abstract
During the late stages of the HIV-1 replication cycle, the viral polyprotein Pr55(Gag) is recruited to the plasma membrane (PM), where it binds phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and directs HIV-1 assembly. We show that Rab27a controls the trafficking of late endosomes carrying phosphatidylinositol 4-kinase type 2 α (PI4KIIα) toward the PM of CD4(+) T cells. Hence, Rab27a promotes high levels of PM phosphatidylinositol 4-phosphate and the localized production of PI(4,5)P2, therefore controlling Pr55(Gag) membrane association. Rab27a also controls PI(4,5)P2 levels at the virus-containing compartments of macrophages. By screening Rab27a effectors, we identified that Slp2a, Slp3, and Slac2b are required for the association of Pr55(Gag) with the PM and that Slp2a cooperates with Rab27a in the recruitment of PI4KIIα to the PM. We conclude that by directing the trafficking of PI4KIIα-positive endosomes toward the PM, Rab27a controls PI(4,5)P2 production and, consequently, HIV-1 replication.
Collapse
Affiliation(s)
- Pehuén Pereyra Gerber
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Mercedes Cabrini
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Carolina Jancic
- Instituto de Medicina Experimental-CONICET, Academia Nacional de Medicina, C1425AUM Buenos Aires, Argentina
| | - Luciana Paoletti
- Instituto de Biologia Molecular y Celular de Rosario-CONICET, S2000EZP Santa Fe, Argentina
| | - Claudia Banchio
- Instituto de Biologia Molecular y Celular de Rosario-CONICET, S2000EZP Santa Fe, Argentina
| | - Catalina von Bilderling
- Instituto de Física de Buenos Aires-CONICET, Departamento de Física; and Centro de Microscopías Avanzadas; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
| | - Lorena Sigaut
- Instituto de Física de Buenos Aires-CONICET, Departamento de Física; and Centro de Microscopías Avanzadas; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
| | - Lía I Pietrasanta
- Instituto de Física de Buenos Aires-CONICET, Departamento de Física; and Centro de Microscopías Avanzadas; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
| | - Gabriel Duette
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Eric O Freed
- Virus-Cell Interaction Section, HIV Drug Resistance Program, National Cancer Institute, Frederick, MD 21702
| | - Genevieve de Saint Basile
- Institut National de la Santé et de la Recherche Médicale U768 and Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, 75015 Paris, France Institut National de la Santé et de la Recherche Médicale U768 and Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, 75015 Paris, France
| | - Catarina Ferreira Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciencia, 2780-156 Oeiras, Portugal
| | - Luis Ferreira Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciencia, 2780-156 Oeiras, Portugal
| | - Sebastian Amigorena
- Centre de Recherche, Institut National de la Santé et de la Recherche Médicale U932, Institut Curie, 75248 Paris, France
| | - Philippe Benaroch
- Centre de Recherche, Institut National de la Santé et de la Recherche Médicale U932, Institut Curie, 75248 Paris, France
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Matías Ostrowski
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
37
|
Lewinski MK, Jafari M, Zhang H, Opella SJ, Guatelli J. Membrane Anchoring by a C-terminal Tryptophan Enables HIV-1 Vpu to Displace Bone Marrow Stromal Antigen 2 (BST2) from Sites of Viral Assembly. J Biol Chem 2015; 290:10919-33. [PMID: 25759385 DOI: 10.1074/jbc.m114.630095] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Indexed: 12/28/2022] Open
Abstract
The restriction factor BST2 (tetherin) prevents the release of enveloped viruses from the host cell and is counteracted by HIV-1 Vpu. Vpu and BST2 interact directly via their transmembrane domains. This interaction enables Vpu to induce the surface down-regulation and the degradation of BST2, but neither of these activities fully accounts for the ability of Vpu to enhance virion release. During a study of naturally occurring Vpu proteins, we found that a tryptophan residue near the Vpu C terminus is particularly important for enhancing virion release. Vpu proteins with a W76G polymorphism degraded and down-regulated BST2 from the cell surface, yet they inefficiently stimulated virion release. Here we explore the mechanism of this anomaly. We find that Trp-76 is critical for the ability of Vpu to displace BST2 from sites of viral assembly in the plane of the plasma membrane. This effect does not appear to involve a general reorganization of the membrane microdomains associated with virion assembly, but rather is a specific effect of Vpu on BST2. Using NMR spectroscopy, we find that the cytoplasmic domain of Vpu and Trp-76 specifically interact with lipids. Moreover, paramagnetic relaxation enhancement studies show that Trp-76 inserts into the lipid. These data are consistent with a model whereby Trp-76 anchors the C terminus of the cytoplasmic tail of Vpu to the plasma membrane, enabling the movement of Vpu-bound BST2 away from viral assembly sites.
Collapse
Affiliation(s)
- Mary K Lewinski
- From the Departments of Medicine and the Veterans Affairs San Diego Healthcare System, San Diego, California 92161
| | | | - Hua Zhang
- Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0679 and
| | - Stanley J Opella
- Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0679 and
| | - John Guatelli
- From the Departments of Medicine and the Veterans Affairs San Diego Healthcare System, San Diego, California 92161
| |
Collapse
|
38
|
Vpu is the main determinant for tetraspanin downregulation in HIV-1-infected cells. J Virol 2015; 89:3247-55. [PMID: 25568205 DOI: 10.1128/jvi.03719-14] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED Tetraspanins constitute a family of cellular proteins that organize various membrane-based processes. Several members of this family, including CD81, are actively recruited by HIV-1 Gag to viral assembly and release sites. Despite their enrichment at viral exit sites, the overall levels of tetraspanins are decreased in HIV-1-infected cells. Here, we identify Vpu as the main viral determinant for tetraspanin downregulation. We also show that reduction of CD81 levels by Vpu is not a by-product of CD4 or BST-2/tetherin elimination from the surfaces of infected cells and likely occurs through an interaction between Vpu and CD81. Finally, we document that Vpu-mediated downregulation of CD81 from the surfaces of infected T cells can contribute to preserving the infectiousness of viral particles, thus revealing a novel Vpu function that promotes virus propagation by modulating the host cell environment. IMPORTANCE The HIV-1 accessory protein Vpu has previously been shown to downregulate various host cell factors, thus helping the virus to overcome restriction barriers, evade immune attack, and maintain the infectivity of viral particles. Our study identifies tetraspanins as an additional group of host factors whose expression at the surfaces of infected cells is lowered by Vpu. While the downregulation of these integral membrane proteins, including CD81 and CD82, likely affects more than one function of HIV-1-infected cells, we document that Vpu-mediated lowering of CD81 levels in viral particles can be critical to maintaining their infectiousness.
Collapse
|
39
|
Chamontin C, Rassam P, Ferrer M, Racine PJ, Neyret A, Lainé S, Milhiet PE, Mougel M. HIV-1 nucleocapsid and ESCRT-component Tsg101 interplay prevents HIV from turning into a DNA-containing virus. Nucleic Acids Res 2014; 43:336-47. [PMID: 25488808 PMCID: PMC4288153 DOI: 10.1093/nar/gku1232] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
HIV-1, the agent of the AIDS pandemic, is an RNA virus that reverse transcribes its RNA genome (gRNA) into DNA, shortly after its entry into cells. Within cells, retroviral assembly requires thousands of structural Gag proteins and two copies of gRNA as well as cellular factors, which converge to the plasma membrane in a finely regulated timeline. In this process, the nucleocapsid domain of Gag (GagNC) ensures gRNA selection and packaging into virions. Subsequent budding and virus release require the recruitment of the cellular ESCRT machinery. Interestingly, mutating GagNC results into the release of DNA-containing viruses, by promo-ting reverse transcription (RTion) prior to virus release, through an unknown mechanism. Therefore, we explored the biogenesis of these DNA-containing particles, combining live-cell total internal-reflection fluorescent microscopy, electron microscopy, trans-complementation assays and biochemical characterization of viral particles. Our results reveal that DNA virus production is the consequence of budding defects associated with Gag aggregation at the plasma membrane and deficiency in the recruitment of Tsg101, a key ESCRT-I component. Indeed, targeting Tsg101 to virus assembly sites restores budding, restricts RTion and favors RNA packaging into viruses. Altogether, our results highlight the role of GagNC in the spatiotemporal control of RTion, via an ESCRT-I-dependent mechanism.
Collapse
Affiliation(s)
- Célia Chamontin
- CPBS, UMR5236 CNRS, University of Montpellier, 34293 Montpellier, France
| | - Patrice Rassam
- Centre de Biochimie Structurale, UMR5048 CNRS, University of Montpellier, 34090 Montpellier, France
| | - Mireia Ferrer
- CPBS, UMR5236 CNRS, University of Montpellier, 34293 Montpellier, France
| | - Pierre-Jean Racine
- CPBS, UMR5236 CNRS, University of Montpellier, 34293 Montpellier, France
| | - Aymeric Neyret
- CPBS, UMR5236 CNRS, University of Montpellier, 34293 Montpellier, France
| | - Sébastien Lainé
- CPBS, UMR5236 CNRS, University of Montpellier, 34293 Montpellier, France
| | - Pierre-Emmanuel Milhiet
- Centre de Biochimie Structurale, UMR5048 CNRS, University of Montpellier, 34090 Montpellier, France U1054 INSERM, 30090 Montpellier, France
| | - Marylène Mougel
- CPBS, UMR5236 CNRS, University of Montpellier, 34293 Montpellier, France
| |
Collapse
|
40
|
Waheed AA, Kuruppu ND, Felton KL, D’Souza D, Freed EO. In COS cells Vpu can both stabilize tetherin expression and counteract its antiviral activity. PLoS One 2014; 9:e111628. [PMID: 25360760 PMCID: PMC4216104 DOI: 10.1371/journal.pone.0111628] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/03/2014] [Indexed: 01/12/2023] Open
Abstract
The interferon-inducible cellular protein tetherin (CD317/BST-2) inhibits the release of a broad range of enveloped viruses. The HIV-1 accessory protein Vpu enhances virus particle release by counteracting this host restriction factor. While the antagonism of human tetherin by Vpu has been associated with both proteasomal and lysosomal degradation, the link between Vpu-mediated tetherin degradation and the ability of Vpu to counteract the antiviral activity of tetherin remains poorly understood. Here, we show that human tetherin is expressed at low levels in African green monkey kidney (COS) cells. However, Vpu markedly increases tetherin expression in this cell line, apparently by sequestering it in an internal compartment that bears lysosomal markers. This stabilization of tetherin by Vpu requires the transmembrane sequence of human tetherin. Although Vpu stabilizes human tetherin in COS cells, it still counteracts the ability of tetherin to suppress virus release. The enhancement of virus release by Vpu in COS cells is associated with a modest reduction in cell-surface tetherin expression, even though the overall expression of tetherin is higher in the presence of Vpu. This study demonstrates that COS cells provide a model system in which Vpu-mediated enhancement of HIV-1 release is uncoupled from Vpu-mediated tetherin degradation.
Collapse
Affiliation(s)
- Abdul A. Waheed
- Virus-Cell Interaction Section, HIV Drug Resistance Program, NCI-Frederick, Frederick, Maryland, United States of America
- * E-mail:
| | - Nishani D. Kuruppu
- Virus-Cell Interaction Section, HIV Drug Resistance Program, NCI-Frederick, Frederick, Maryland, United States of America
| | - Kathryn L. Felton
- Virus-Cell Interaction Section, HIV Drug Resistance Program, NCI-Frederick, Frederick, Maryland, United States of America
| | - Darren D’Souza
- Virus-Cell Interaction Section, HIV Drug Resistance Program, NCI-Frederick, Frederick, Maryland, United States of America
| | - Eric O. Freed
- Virus-Cell Interaction Section, HIV Drug Resistance Program, NCI-Frederick, Frederick, Maryland, United States of America
| |
Collapse
|
41
|
Basic motifs target PSGL-1, CD43, and CD44 to plasma membrane sites where HIV-1 assembles. J Virol 2014; 89:454-67. [PMID: 25320329 DOI: 10.1128/jvi.02178-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED HIV-1 incorporates various host membrane proteins during particle assembly at the plasma membrane; however, the mechanisms mediating this incorporation process remain poorly understood. We previously showed that the HIV-1 structural protein Gag localizes to the uropod, a rear-end structure of polarized T cells, and that assembling Gag copatches with a subset, but not all, of the uropod-directed proteins, i.e., PSGL-1, CD43, and CD44, in nonpolarized T cells. The latter observation suggests the presence of a mechanism promoting virion incorporation of these cellular proteins. To address this possibility and identify molecular determinants, in the present study we examined coclustering between Gag and the transmembrane proteins in T and HeLa cells using quantitative two-color superresolution localization microscopy. Consistent with the findings of the T-cell copatching study, we found that basic residues within the matrix domain of Gag are required for Gag-PSGL-1 coclustering. Notably, the presence of a polybasic sequence in the PSGL-1 cytoplasmic domain significantly enhanced this coclustering. We also found that polybasic motifs present in the cytoplasmic tails of CD43 and CD44 also promote their coclustering with Gag. ICAM-1 and ICAM-3, uropod-directed proteins that do not copatch with Gag in T cells, and CD46, a non-uropod-directed protein, showed no or little coclustering with Gag. However, replacing their cytoplasmic tails with the cytoplasmic tail of PSGL-1 significantly enhanced their coclustering with Gag. Altogether, these results identify a novel mechanism for host membrane protein association with assembling HIV-1 Gag in which polybasic sequences present in the cytoplasmic tails of the membrane proteins and in Gag are the major determinants. IMPORTANCE Nascent HIV-1 particles incorporate many host plasma membrane proteins during assembly. However, it is largely unknown what mechanisms promote the association of these proteins with virus assembly sites within the plasma membrane. Notably, our previous study showed that HIV-1 structural protein Gag colocalizes with a group of uropod-directed transmembrane proteins, PSGL-1, CD43, and CD44, at the plasma membrane of T cells. The results obtained in the current study using superresolution localization microscopy suggest the presence of a novel molecular mechanism promoting the association of PSGL-1, CD43, and CD44 with assembling HIV-1 which relies on polybasic sequences in HIV-1 Gag and in cytoplasmic domains of the transmembrane proteins. This information advances our understanding of virion incorporation of host plasma membrane proteins, some of which modulate virus spread positively or negatively, and suggests a possible new strategy to enrich HIV-1-based lentiviral vectors with a desired transmembrane protein.
Collapse
|
42
|
Abstract
Tetraspanins are a family of proteins with four transmembrane domains that play a role in many aspects of cell biology and physiology; they are also used by several pathogens for infection and regulate cancer progression. Many tetraspanins associate specifically and directly with a limited number of proteins, and also with other tetraspanins, thereby generating a hierarchical network of interactions. Through these interactions, tetraspanins are believed to have a role in cell and membrane compartmentalization. In this Cell Science at a Glance article and the accompanying poster, we describe the basic principles underlying tetraspanin-based assemblies and highlight examples of how tetraspanins regulate the trafficking and function of their partner proteins that are required for the normal development and function of several organs, including, in humans, the eye, the kidney and the immune system.
Collapse
Affiliation(s)
- Stéphanie Charrin
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| | - Stéphanie Jouannet
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| | - Claude Boucheix
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| | - Eric Rubinstein
- Inserm, U1004, F-94807, Villejuif, France Université Paris-Sud, Institut André Lwoff, F-94807 Villejuif, France
| |
Collapse
|
43
|
Mariani C, Desdouits M, Favard C, Benaroch P, Muriaux DM. Role of Gag and lipids during HIV-1 assembly in CD4(+) T cells and macrophages. Front Microbiol 2014; 5:312. [PMID: 25009540 PMCID: PMC4069574 DOI: 10.3389/fmicb.2014.00312] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 06/08/2014] [Indexed: 12/25/2022] Open
Abstract
HIV-1 is an RNA enveloped virus that preferentially infects CD4+ T lymphocytes and also macrophages. In CD4+ T cells, HIV-1 mainly buds from the host cell plasma membrane. The viral Gag polyprotein targets the plasma membrane and is the orchestrator of the HIV assembly as its expression is sufficient to promote the formation of virus-like particles carrying a lipidic envelope derived from the host cell membrane. Certain lipids are enriched in the viral membrane and are thought to play a key role in the assembly process and the envelop composition. A large body of work performed on infected CD4+ T cells has provided important knowledge about the assembly process and the membrane virus lipid composition. While HIV assembly and budding in macrophages is thought to follow the same general Gag-driven mechanism as in T-lymphocytes, the HIV cycle in macrophage exhibits specific features. In these cells, new virions bud from the limiting membrane of seemingly intracellular compartments, where they accumulate while remaining infectious. These structures are now often referred to as Virus Containing Compartments (VCCs). Recent studies suggest that VCCs represent intracellularly sequestered regions of the plasma membrane, but their precise nature remains elusive. The proteomic and lipidomic characterization of virions produced by T cells or macrophages has highlighted the similarity between their composition and that of the plasma membrane of producer cells, as well as their enrichment in acidic lipids, some components of raft lipids and in tetraspanin-enriched microdomains. It is likely that Gag promotes the coalescence of these components into an assembly platform from which viral budding takes place. How Gag exactly interacts with membrane lipids and what are the mechanisms involved in the interaction between the different membrane nanodomains within the assembly platform remains unclear. Here we review recent literature regarding the role of Gag and lipids on HIV-1 assembly in CD4+ T cells and macrophages.
Collapse
Affiliation(s)
- Charlotte Mariani
- Membrane Domains and Viral Assembly, CNRS UMR-5236, Centre d'étude d'agents Pathogènes et Biotechnologies pour la Santé Montpellier, Cedex, France
| | - Marion Desdouits
- Intracellular Transport and Immunity, Immunité et Cancer, Institut Curie - Inserm U932 Paris, France
| | - Cyril Favard
- Membrane Domains and Viral Assembly, CNRS UMR-5236, Centre d'étude d'agents Pathogènes et Biotechnologies pour la Santé Montpellier, Cedex, France
| | - Philippe Benaroch
- Intracellular Transport and Immunity, Immunité et Cancer, Institut Curie - Inserm U932 Paris, France
| | - Delphine M Muriaux
- Membrane Domains and Viral Assembly, CNRS UMR-5236, Centre d'étude d'agents Pathogènes et Biotechnologies pour la Santé Montpellier, Cedex, France
| |
Collapse
|
44
|
Ehrlich LS, Medina GN, Photiadis S, Whittredge PB, Watanabe S, Taraska JW, Carter CA. Tsg101 regulates PI(4,5)P2/Ca(2+) signaling for HIV-1 Gag assembly. Front Microbiol 2014; 5:234. [PMID: 24904548 PMCID: PMC4033031 DOI: 10.3389/fmicb.2014.00234] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 04/30/2014] [Indexed: 11/13/2022] Open
Abstract
Our previous studies identified the 1,4,5-inositol trisphosphate receptor (IP3R), a channel mediating release of Ca2+ from ER stores, as a cellular factor differentially associated with HIV-1 Gag that might facilitate ESCRT function in virus budding. Channel opening requires activation that is initiated by binding of 1,4,5-triphosphate (IP3), a product of phospholipase C (PLC)-mediated PI(4,5)P2 hydrolysis. The store emptying that follows stimulates store refilling which requires intact PI(4,5)P2. Raising cytosolic Ca2+ promotes viral particle production and our studies indicate that IP3R and the ER Ca2+ store are the physiological providers of Ca2+ for Gag assembly and release. Here, we show that Gag modulates ER store gating and refilling. Cells expressing Gag exhibited a higher cytosolic Ca2+ level originating from the ER store than control cells, suggesting that Gag induced release of store Ca2+. This property required the PTAP motif in Gag that recruits Tsg101, an ESCRT-1 component. Consistent with cytosolic Ca2+ elevation, Gag accumulation at the plasma membrane was found to require continuous IP3R activation. Like other IP3R channel modulators, Gag was detected in physical proximity to the ER and to endogenous IP3R, as indicated respectively by total internal reflection fluorescence (TIRF) and immunoelectron microscopy (IEM) or indirect immunofluorescence. Reciprocal co-immunoprecipitation suggested that Gag and IP3R proximity is favored when the PTAP motif in Gag is intact. Gag expression was also accompanied by increased PI(4,5)P2 accumulation at the plasma membrane, a condition favoring store refilling capacity. Supporting this notion, Gag particle production was impervious to treatment with 2-aminoethoxydiphenyl borate, an inhibitor of a refilling coupling interaction. In contrast, particle production by a Gag mutant lacking the PTAP motif was reduced. We conclude that a functional PTAP L domain, and by inference Tsg101 binding, confers Gag with an ability to modulate both ER store Ca2+ release and ER store refilling.
Collapse
Affiliation(s)
- Lorna S Ehrlich
- Molecular Genetics and Microbiology, Stony Brook University Stony Brook, NY, USA
| | - Gisselle N Medina
- Molecular Genetics and Microbiology, Stony Brook University Stony Brook, NY, USA
| | - Sara Photiadis
- Molecular Genetics and Microbiology, Stony Brook University Stony Brook, NY, USA
| | - Paul B Whittredge
- Laboratory of Molecular Biophysics, National Heart Lung and Blood Institute, National Institutes of Health Bethesda, MD, USA
| | - Susan Watanabe
- Molecular Genetics and Microbiology, Stony Brook University Stony Brook, NY, USA
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart Lung and Blood Institute, National Institutes of Health Bethesda, MD, USA
| | - Carol A Carter
- Molecular Genetics and Microbiology, Stony Brook University Stony Brook, NY, USA
| |
Collapse
|
45
|
Yandrapalli N, Muriaux D, Favard C. Lipid domains in HIV-1 assembly. Front Microbiol 2014; 5:220. [PMID: 24904536 PMCID: PMC4033000 DOI: 10.3389/fmicb.2014.00220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/26/2014] [Indexed: 12/14/2022] Open
Abstract
In CD+4 T cells, HIV-1 buds from the host cell plasma membrane. The viral Gag polyprotein is mainly responsible for this process. However, the intimate interaction of Gag and lipids at the plasma membrane as well as its consequences, in terms of lipids lateral organization and virus assembly, is still under debate. In this review we propose to revisit the role of plasma membrane lipids in HIV-1 Gag targeting and assembly, at the light of lipid membranes biophysics and literature dealing with Gag-lipid interactions.
Collapse
Affiliation(s)
- Naresh Yandrapalli
- Centre d'étude des Pathogènes et de Biotechnologies pour la Santé, CNRS UMR-5236 Montpellier Cedex, France
| | - Delphine Muriaux
- Centre d'étude des Pathogènes et de Biotechnologies pour la Santé, CNRS UMR-5236 Montpellier Cedex, France
| | - Cyril Favard
- Centre d'étude des Pathogènes et de Biotechnologies pour la Santé, CNRS UMR-5236 Montpellier Cedex, France
| |
Collapse
|
46
|
Dahmane S, Rubinstein E, Milhiet PE. Viruses and tetraspanins: lessons from single molecule approaches. Viruses 2014; 6:1992-2011. [PMID: 24800676 PMCID: PMC4036545 DOI: 10.3390/v6051992] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/24/2014] [Accepted: 04/10/2014] [Indexed: 12/15/2022] Open
Abstract
Tetraspanins are four-span membrane proteins that are widely distributed in multi-cellular organisms and involved in several infectious diseases. They have the unique property to form a network of protein-protein interaction within the plasma membrane, due to the lateral associations with one another and with other membrane proteins. Tracking tetraspanins at the single molecule level using fluorescence microscopy has revealed the membrane behavior of the tetraspanins CD9 and CD81 in epithelial cell lines, providing a first dynamic view of this network. Single molecule tracking highlighted that these 2 proteins can freely diffuse within the plasma membrane but can also be trapped, permanently or transiently, in tetraspanin-enriched areas. More recently, a similar strategy has been used to investigate tetraspanin membrane behavior in the context of human immunodeficiency virus type 1 (HIV-1) and hepatitis C virus (HCV) infection. In this review we summarize the main results emphasizing the relationship in terms of membrane partitioning between tetraspanins, some of their partners such as Claudin-1 and EWI-2, and viral proteins during infection. These results will be analyzed in the context of other membrane microdomains, stressing the difference between raft and tetraspanin-enriched microdomains, but also in comparison with virus diffusion at the cell surface. New advanced single molecule techniques that could help to further explore tetraspanin assemblies will be also discussed.
Collapse
Affiliation(s)
- Selma Dahmane
- Inserm, Unité 1054, Single Molecule Biophysics Department, Centre de Biochimie Structurale, 34090, Montpellier, France.
| | | | - Pierre-Emmanuel Milhiet
- Inserm, Unité 1054, Single Molecule Biophysics Department, Centre de Biochimie Structurale, 34090, Montpellier, France.
| |
Collapse
|
47
|
Rocha-Perugini V, Gordon-Alonso M, Sánchez-Madrid F. PIP2: choreographer of actin-adaptor proteins in the HIV-1 dance. Trends Microbiol 2014; 22:379-88. [PMID: 24768560 DOI: 10.1016/j.tim.2014.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/11/2014] [Accepted: 03/25/2014] [Indexed: 02/06/2023]
Abstract
The actin cytoskeleton plays a key role during the replication cycle of human immunodeficiency virus-1 (HIV-1). HIV-1 infection is affected by cellular proteins that influence the clustering of viral receptors or the subcortical actin cytoskeleton. Several of these actin-adaptor proteins are controlled by the second messenger phosphatidylinositol 4,5-biphosphate (PIP2), an important regulator of actin organization. PIP2 production is induced by HIV-1 attachment and facilitates viral infection. However, the importance of PIP2 in regulating cytoskeletal proteins and thus HIV-1 infection has been overlooked. This review examines recent reports describing the roles played by actin-adaptor proteins during HIV-1 infection of CD4+ T cells, highlighting the influence of the signaling lipid PIP2 in this process.
Collapse
Affiliation(s)
- Vera Rocha-Perugini
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain; Vascular Biology and Inflammation Department, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Mónica Gordon-Alonso
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain; Vascular Biology and Inflammation Department, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
| |
Collapse
|
48
|
Gregory DA, Olinger GY, Lucas TM, Johnson MC. Diverse viral glycoproteins as well as CD4 co-package into the same human immunodeficiency virus (HIV-1) particles. Retrovirology 2014; 11:28. [PMID: 24708808 PMCID: PMC3985584 DOI: 10.1186/1742-4690-11-28] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 03/19/2014] [Indexed: 12/17/2022] Open
Abstract
Background Retroviruses can acquire not only their own glycoproteins as they bud from the cellular membrane, but also some cellular and foreign viral glycoproteins. Many of these non-native glycoproteins are actively recruited to budding virions, particularly other viral glycoproteins. This observation suggests that there may be a conserved mechanism underlying the recruitment of glycoproteins into viruses. If a conserved mechanism is used, diverse glycoproteins should localize to a single budding retroviral particle. On the other hand, if viral glycoproteins have divergent mechanisms for recruitment, the different glycoproteins could segregate into different particles. Results To determine if co-packaging occurs among different glycoproteins, we designed an assay that combines virion antibody capture and a determination of infectivity based on a luciferase reporter. Virions were bound to a plate with an antibody against one glycoprotein, and then the infectivity was measured with cells that allow entry only with a second glycoprotein. We tested pairings of glycoproteins from HIV, murine leukemia virus (MLV), Rous sarcoma virus (RSV), vesicular stomatitis virus (VSV), and Ebola virus. The results showed that glycoproteins that were actively recruited into virions were co-packaged efficiently with each other. We also tested cellular proteins and found CD4 also had a similar correlation between active recruitment and efficient co-packaging, but other cellular proteins did not. Conclusion Glycoproteins that are actively incorporated into HIV-1 virions are efficiently co-packaged into the same virus particles, suggesting that the same general mechanism for recruitment may act in many viruses.
Collapse
Affiliation(s)
| | | | | | - Marc C Johnson
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
49
|
Grove J. Super-resolution microscopy: a virus' eye view of the cell. Viruses 2014; 6:1365-78. [PMID: 24651030 PMCID: PMC3970155 DOI: 10.3390/v6031365] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/01/2014] [Accepted: 03/11/2014] [Indexed: 12/24/2022] Open
Abstract
It is difficult to observe the molecular choreography between viruses and host cell components, as they exist on a spatial scale beyond the reach of conventional microscopy. However, novel super-resolution microscopy techniques have cast aside technical limitations to reveal a nanoscale view of virus replication and cell biology. This article provides an introduction to super-resolution imaging; in particular, localisation microscopy, and explores the application of such technologies to the study of viruses and tetraspanins, the topic of this special issue.
Collapse
Affiliation(s)
- Joe Grove
- Institute of Immunity and Transplantation, University College London, London NW3 2PF, UK.
| |
Collapse
|
50
|
Symeonides M, Lambelé M, Roy NH, Thali M. Evidence showing that tetraspanins inhibit HIV-1-induced cell-cell fusion at a post-hemifusion stage. Viruses 2014; 6:1078-90. [PMID: 24608085 PMCID: PMC3970140 DOI: 10.3390/v6031078] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/14/2014] [Accepted: 02/20/2014] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) transmission takes place primarily through cell-cell contacts known as virological synapses. Formation of these transient adhesions between infected and uninfected cells can lead to transmission of viral particles followed by separation of the cells. Alternatively, the cells can fuse, thus forming a syncytium. Tetraspanins, small scaffolding proteins that are enriched in HIV-1 virions and actively recruited to viral assembly sites, have been found to negatively regulate HIV-1 Env-induced cell-cell fusion. How these transmembrane proteins inhibit membrane fusion, however, is currently not known. As a first step towards elucidating the mechanism of fusion repression by tetraspanins, e.g., CD9 and CD63, we sought to identify the stage of the fusion process during which they operate. Using a chemical epistasis approach, four fusion inhibitors were employed in tandem with CD9 overexpression. Cells overexpressing CD9 were found to be sensitized to inhibitors targeting the pre-hairpin and hemifusion intermediates, while they were desensitized to an inhibitor of the pore expansion stage. Together with the results of a microscopy-based dye transfer assay, which revealed CD9- and CD63-induced hemifusion arrest, our investigations strongly suggest that tetraspanins block HIV-1-induced cell-cell fusion at the transition from hemifusion to pore opening.
Collapse
Affiliation(s)
- Menelaos Symeonides
- Graduate Program in Cell and Molecular Biology, University of Vermont, Burlington, VT 05405, USA.
| | - Marie Lambelé
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA.
| | - Nathan H Roy
- Graduate Program in Cell and Molecular Biology, University of Vermont, Burlington, VT 05405, USA.
| | - Markus Thali
- Graduate Program in Cell and Molecular Biology, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|