1
|
Cappe B, Vandenabeele P, Riquet FB. A guide to the expanding field of extracellular vesicles and their release in regulated cell death programs. FEBS J 2024; 291:2068-2090. [PMID: 37872002 DOI: 10.1111/febs.16981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Homeostasis disruption is visible at the molecular and cellular levels and may often lead to cell death. This vital process allows us to maintain the more extensive system's integrity by keeping the different features (genetic, metabolic, physiologic, and individual) intact. Interestingly, while cells can die in different manners, dying cells still communicate with their environment. This communication was, for a long time, perceived as only driven by the release of soluble factors. However, it has now been reconsidered with the increasing interest in extracellular vesicles (EVs), which are discovered to be released during different regulated cell death programs, with the observation of specific effects. EVs are game changers in the paradigm of cell-cell communication with tremendous implications in fundamental research with regard to noncell autonomous functions, as well as in biomarkers research, all of which are geared toward diagnostic and therapeutic purposes. This review is composed of two main parts. The first is a comprehensive presentation of the state of the art of the EV field at large. In the second part, we focus on EVs discovered to be released during different regulated cell death programs, also known as cell death EVs (cdEVs), and EV-associated specific effects on recipient cells in the context of cell death and inflammation/inflammatory responses.
Collapse
Affiliation(s)
- Benjamin Cappe
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Franck B Riquet
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
- University of Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, France
| |
Collapse
|
2
|
The large extracellular loop of CD63 interacts with gp41 of HIV-1 and is essential for establishing the virological synapse. Sci Rep 2021; 11:10011. [PMID: 33976357 PMCID: PMC8113602 DOI: 10.1038/s41598-021-89523-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/21/2021] [Indexed: 11/09/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) persists lifelong in infected individuals and has evolved unique strategies in order to evade the immune system. One of these strategies is the direct cell-to-cell spread of HIV-1. The formation of a virological synapse (VS) between donor and target cell is important for this process. Tetraspanins are cellular proteins that are actively involved in the formation of a VS. However, the molecular mechanisms of recruiting host proteins for the cell–cell transfer of particles to the VS remains unclear. Our study has mapped the binding site for the transmembrane envelope protein gp41 of HIV-1 within the large extracellular loop (LEL) of CD63 and showed that this interaction occurs predominantly at the VS between T cells where viral particles are transferred. Mutations within the highly conserved CCG motif of the tetraspanin superfamily abrogated recruiting of expressed HIV-1 GFP fused Gag core protein and CD63 to the VS. This demonstrates the biological significance of CD63 for enhanced formation of a VS. Since cell–cell spread of HIV-1 is a major route of persistent infection, these results highlight the central role of CD63 as a member of the tetraspanin superfamily during HIV-1 infection and pathogenesis.
Collapse
|
3
|
Transfiguracion J, Tran MY, Lanthier S, Tremblay S, Coulombe N, Acchione M, Kamen AA. Rapid In-Process Monitoring of Lentiviral Vector Particles by High-Performance Liquid Chromatography. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:803-810. [PMID: 32953931 PMCID: PMC7479275 DOI: 10.1016/j.omtm.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/31/2020] [Indexed: 12/28/2022]
Abstract
Lentiviral vectors (LVs) are a popular gene delivery tool in cell and gene therapy and they are a primary tool for ex vivo transduction of T cells for expression of chimeric antigen receptor (CAR) in CAR-T cell therapies. Extensive process and product characterization are required in manufacturing virus-based gene vectors to better control batch-to-batch variability. However, it has been an ongoing challenge to make quantitative assessments of LV product because current analytical tools often are low throughput and lack robustness and standardization is still required. This paper presents a high-throughput and robust physico-chemical characterization method that directly assesses total LV particles. With simple sample preparation and fast elution time (6.24 min) of the LV peak in 440 mM NaCl (in 20 mM Tris-HCl [pH 7.5]), this ion exchange high-performance liquid chromatography (IEX-HPLC) method is ideal for routine in-process monitoring to facilitate the development of scalable and robust LV manufacturing processes. Furthermore, this HPLC method is suitable for the analysis of all in-process samples, from crude samples such as LV supernatants to final purified products. The linearity range of the standard curve is 3.13 × 108 to 1.0 × 1010 total particles/mL, and both the intra- and inter-assay variabilities are less than 5%.
Collapse
Affiliation(s)
- Julia Transfiguracion
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Michelle Yen Tran
- Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, 817 Sherbrooke West #270, Montreal, QC H3A 0C3, Canada
| | - Stéphane Lanthier
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Sonia Tremblay
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Nathalie Coulombe
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Mauro Acchione
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Amine A Kamen
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada.,Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, 817 Sherbrooke West #270, Montreal, QC H3A 0C3, Canada
| |
Collapse
|
4
|
Watanabe Y, Bowden TA, Wilson IA, Crispin M. Exploitation of glycosylation in enveloped virus pathobiology. Biochim Biophys Acta Gen Subj 2019; 1863:1480-1497. [PMID: 31121217 PMCID: PMC6686077 DOI: 10.1016/j.bbagen.2019.05.012] [Citation(s) in RCA: 318] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022]
Abstract
Glycosylation is a ubiquitous post-translational modification responsible for a multitude of crucial biological roles. As obligate parasites, viruses exploit host-cell machinery to glycosylate their own proteins during replication. Viral envelope proteins from a variety of human pathogens including HIV-1, influenza virus, Lassa virus, SARS, Zika virus, dengue virus, and Ebola virus have evolved to be extensively glycosylated. These host-cell derived glycans facilitate diverse structural and functional roles during the viral life-cycle, ranging from immune evasion by glycan shielding to enhancement of immune cell infection. In this review, we highlight the imperative and auxiliary roles glycans play, and how specific oligosaccharide structures facilitate these functions during viral pathogenesis. We discuss the growing efforts to exploit viral glycobiology in the development of anti-viral vaccines and therapies.
Collapse
Affiliation(s)
- Yasunori Watanabe
- School of Biological Sciences and Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK; Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford OX3 7BN, UK; Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Thomas A Bowden
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford OX3 7BN, UK
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Max Crispin
- School of Biological Sciences and Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
5
|
The duck EB66® cell substrate reveals a novel retrotransposon. Biologicals 2019; 61:22-31. [DOI: 10.1016/j.biologicals.2019.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 11/18/2022] Open
|
6
|
Ressel S, Rosca A, Gordon K, Buck AH. Extracellular RNA in viral-host interactions: Thinking outside the cell. WILEY INTERDISCIPLINARY REVIEWS. RNA 2019; 10:e1535. [PMID: 30963709 PMCID: PMC6617787 DOI: 10.1002/wrna.1535] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/15/2022]
Abstract
Small RNAs and their associated RNA interference (RNAi) pathways underpin diverse mechanisms of gene regulation and genome defense across all three kingdoms of life and are integral to virus-host interactions. In plants, fungi and many animals, an ancestral RNAi pathway exists as a host defense mechanism whereby viral double-stranded RNA is processed to small RNAs that enable recognition and degradation of the virus. While this antiviral RNAi pathway is not generally thought to be present in mammals, other RNAi mechanisms can influence infection through both viral- and host-derived small RNAs. Furthermore, a burgeoning body of data suggests that small RNAs in mammals can function in a non-cell autonomous manner to play various roles in cell-to-cell communication and disease through their transport in extracellular vesicles. While vesicular small RNAs have not been proposed as an antiviral defense pathway per se, there is increasing evidence that the export of host- or viral-derived RNAs from infected cells can influence various aspects of the infection process. This review discusses the current knowledge of extracellular RNA functions in viral infection and the technical challenges surrounding this field of research. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs RNA in Disease and Development > RNA in Disease Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Sarah Ressel
- Institute of Immunology and Infection Research, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Adelina Rosca
- Department of VirologyCarol Davila University of Medicine and PharmacyBucharestRomania
| | - Katrina Gordon
- Institute of Immunology and Infection Research, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Amy H. Buck
- Institute of Immunology and Infection Research, School of Biological SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
7
|
Reiter K, Aguilar PP, Wetter V, Steppert P, Tover A, Jungbauer A. Separation of virus-like particles and extracellular vesicles by flow-through and heparin affinity chromatography. J Chromatogr A 2019; 1588:77-84. [DOI: 10.1016/j.chroma.2018.12.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 12/31/2022]
|
8
|
Burnie J, Guzzo C. The Incorporation of Host Proteins into the External HIV-1 Envelope. Viruses 2019; 11:v11010085. [PMID: 30669528 PMCID: PMC6356245 DOI: 10.3390/v11010085] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
The incorporation of biologically active host proteins into HIV-1 is a well-established phenomenon, particularly due to the budding mechanism of viral egress in which viruses acquire their external lipid membrane directly from the host cell. While this mechanism might seemingly imply that host protein incorporation is a passive uptake of all cellular antigens associated with the plasma membrane at the site of budding, this is not the case. Herein, we review the evidence indicating that host protein incorporation can be a selective and conserved process. We discuss how HIV-1 virions displaying host proteins on their surface can exhibit a myriad of altered phenotypes, with notable impacts on infectivity, homing, neutralization, and pathogenesis. This review describes the canonical and emerging methods to detect host protein incorporation, highlights the well-established host proteins that have been identified on HIV-1 virions, and reflects on the role of these incorporated proteins in viral pathogenesis and therapeutic targeting. Despite many advances in HIV treatment and prevention, there remains a global effort to develop increasingly effective anti-HIV therapies. Given the broad range of biologically active host proteins acquired on the surface of HIV-1, additional studies on the mechanisms and impacts of these incorporated host proteins may inform the development of novel treatments and vaccine designs.
Collapse
Affiliation(s)
- Jonathan Burnie
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| | - Christina Guzzo
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| |
Collapse
|
9
|
Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 2019; 21:9-17. [PMID: 30602770 DOI: 10.1038/s41556-018-0250-9] [Citation(s) in RCA: 2303] [Impact Index Per Article: 460.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023]
Abstract
The ability of exosomes to transfer cargo from donor to acceptor cells, thereby triggering phenotypic changes in the latter, has generated substantial interest in the scientific community. However, the extent to which exosomes differ from other extracellular vesicles in terms of their biogenesis and functions remains ill-defined. Here, we discuss the current knowledge on the specificities of exosomes and other types of extracellular vesicles, and their roles as important agents of cell-to-cell communication.
Collapse
|
10
|
Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 2019. [PMID: 30602770 DOI: 10.1038/s41556-018-0250-259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2023]
Abstract
The ability of exosomes to transfer cargo from donor to acceptor cells, thereby triggering phenotypic changes in the latter, has generated substantial interest in the scientific community. However, the extent to which exosomes differ from other extracellular vesicles in terms of their biogenesis and functions remains ill-defined. Here, we discuss the current knowledge on the specificities of exosomes and other types of extracellular vesicles, and their roles as important agents of cell-to-cell communication.
Collapse
Affiliation(s)
- Mathilde Mathieu
- Institut Curie, PSL Research University, INSERM U932, Paris, France
- Université Paris Descartes, Paris, France
| | | | - Grégory Lavieu
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932, Paris, France.
| |
Collapse
|
11
|
Sviben D, Forcic D, Halassy B, Allmaier G, Marchetti-Deschmann M, Brgles M. Mass spectrometry-based investigation of measles and mumps virus proteome. Virol J 2018; 15:160. [PMID: 30326905 PMCID: PMC6192076 DOI: 10.1186/s12985-018-1073-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/02/2018] [Indexed: 02/08/2023] Open
Abstract
Background Measles (MEV) and mumps virus (MUV) are enveloped, non-segmented, negative single stranded RNA viruses of the family Paramyxoviridae, and are the cause of measles and mumps, respectively, both preventable by vaccination. Aside from proteins coded by the viral genome, viruses are considered to contain host cell proteins (HCPs). The presence of extracellular vesicles (ECVs), which are often co-purified with viruses due to their similarity in size, density and composition, also contributes to HCPs detected in virus preparations, and this has often been neglected. The aim was to identify which virus-coded proteins are present in MEV and MUV virions, and to try to detect which HCPs, if any, are incorporated inside the virions or adsorbed on their outer surface, and which are more likely to be a contamination from co-purified ECVs. Methods MUV, MEV and ECVs were purified by ultracentrifugation, hydrophobic interaction chromatography and immunoaffinity chromatography, proteins in the samples were resolved by SDS-PAGE and subjected to identification by MALDI-TOF/TOF-MS. A comparative analysis of HCPs present in all samples was carried out. Results By proteomics approach, it was verified that almost all virus-coded proteins are present in MEV and MUV particles. Protein C in MEV which was until now considered to be non-structural viral protein, was found to be present inside the MeV virions. Results on the presence of HCPs in differently purified virus preparations imply that actin, annexins, cyclophilin A, moesin and integrin β1 are part of the virions. Conclusions All HCPs detected in the viruses are present in ECVs as well, indicating their possible function in vesicle formation, or that most of them are only present in ECVs. Only five HCPs were constantly present in purified virus preparations, regardless of the purification method used, implying they are likely the integral part of the virions. The approach described here is helpful for further investigation of HCPs in other virus preparations. Electronic supplementary material The online version of this article (10.1186/s12985-018-1073-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dora Sviben
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia. .,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia.
| | - Dubravko Forcic
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Günter Allmaier
- Institute of Chemical Technologies and Analytics, TU Wien, Getreidemarkt 9, AT-1060, Vienna, Austria
| | | | - Marija Brgles
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| |
Collapse
|
12
|
Barklis E, Staubus AO, Mack A, Harper L, Barklis RL, Alfadhli A. Lipid biosensor interactions with wild type and matrix deletion HIV-1 Gag proteins. Virology 2018; 518:264-271. [PMID: 29549788 DOI: 10.1016/j.virol.2018.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 11/19/2022]
Abstract
The matrix (MA) domain of the HIV-1 precursor Gag protein (PrGag) has been shown interact with the HIV-1 envelope (Env) protein, and to direct PrGag proteins to plasma membrane (PM) assembly sites by virtue of its affinity to phosphatidylinositol-4,5-bisphosphate (PI[4,5]P2). Unexpectedly, HIV-1 viruses with large MA deletions (ΔMA) have been shown to be conditionally infectious as long as they are matched with Env truncation mutant proteins or alternative viral glycoproteins. To characterize the interactions of wild type (WT) and ΔMA Gag proteins with PI(4,5)P2 and other acidic phospholipids, we have employed a set of lipid biosensors as probes. Our investigations showed marked differences in WT and ΔMA Gag colocalization with biosensors, effects on biosensor release, and association of biosensors with virus-like particles. These results demonstrate an alternative approach to the analysis of viral protein-lipid associations, and provide new data as to the lipid compositions of HIV-1 assembly sites.
Collapse
Affiliation(s)
- Eric Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States.
| | - August O Staubus
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Andrew Mack
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Logan Harper
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Robin Lid Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Ayna Alfadhli
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| |
Collapse
|
13
|
Exosomes Mediate Intercellular Transmission of Porcine Reproductive and Respiratory Syndrome Virus. J Virol 2018; 92:JVI.01734-17. [PMID: 29187541 DOI: 10.1128/jvi.01734-17] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/22/2017] [Indexed: 12/13/2022] Open
Abstract
Exosomes are small membrane-enclosed vesicles produced by various cells and actively released into the extracellular space. They participate in intercellular communication and transfer of biologically active proteins, lipids, and nucleic acids. Accumulating evidence suggests that exosomes derived from cells infected by some viruses selectively encapsulate viral proteins, genetic materials, or even virions to mediate cell-to-cell communication and/or virus transmission. Porcine reproductive and respiratory syndrome virus (PRRSV) is an Arterivirus that has been devastating the global swine industry since the late 1980s. Recent studies have shown that major proteins secreted from PRRSV-infected cells are exosomal proteins and that the serum-derived exosomes from PRRSV-infected pigs contain viral proteins. However, the role of exosomes in PRRSV infection remains unclear. In this study, purified exosomes isolated from PRRSV-infected cells were shown with reverse transcription-PCR and mass spectrometry to contain viral genomic RNA and partial viral proteins. Furthermore, exosomes from PRRSV-infected cells established productive infection in both PRRSV-susceptible and -nonsusceptible cells. More importantly, exosome-mediated infection was not completely blocked by PRRSV-specific neutralizing antibodies. In summary, this study demonstrated that exosomes can mediate PRRSV transmission and are even resistant to antibody neutralization, identifying a potential immune evasion mechanism utilized by PRRSV.IMPORTANCE Exosomes have recently been characterized as bioactive vesicles that function to promote intercellular communication. The exosomes from virally infected cells containing altered compositions confer numerous novel functionalities. A study of the secretome of cells infected with PRRSV indicated that the exosomal pathway is strongly activated by PRRSV infection. Here, we demonstrate that PRRSV can utilize host exosomes to infect naive healthy cells. Furthermore, exosome-mediated viral transmission is largely resistant to PRRSV-specific neutralizing antibodies. Our study provides novel insights into an alternative mechanism of PRRSV transmission that can compromise the host's anti-PRRSV immune response.
Collapse
|
14
|
Identification of HIV-1-Based Virus-like Particles by Multifrequency Atomic Force Microscopy. Biophys J 2017; 111:1173-1179. [PMID: 27653476 DOI: 10.1016/j.bpj.2016.07.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/05/2016] [Accepted: 07/11/2016] [Indexed: 11/21/2022] Open
Abstract
Virus-like particles (VLPs) have become a promising platform for vaccine production. VLPs are formed by structural viral proteins that inherently self-assemble when expressed in a host cell. They represent a highly immunogenic and safe vaccine platform, due to the absence of the viral genome and its high protein density. One of the most important parameters in vaccine production is the quality of the product. A related bottleneck in VLP-based products is the presence of cellular vesicles as a major contaminant in the preparations, which will require the set up of techniques allowing for specific discrimination of VLPs from host vesicular bodies. In this work novel, to our knowledge, multifrequency (MF) atomic force microscopy (AFM) has permitted full structural nanophysical characterization by its access to the virus capsid of the HIV-based VLPs. The assessment of these particles by advanced amplitude modulation-frequency modulation (AM-FM) viscoelastic mapping mode has enhanced the imaging resolution of their nanomechanical properties, opening a new window for the study of the biophysical attributes of VLPs. Finally, the identification and differentiation of HIV-based VLPs from cellular vesicles has been performed under ambient conditions, providing, to our knowledge, novel methodology for the monitoring and quality control of VLPs.
Collapse
|
15
|
Kondratov KA, Petrova TA, Mikhailovskii VY, Ivanova AN, Kostareva AA, Fedorov AV. A study of extracellular vesicles isolated from blood plasma conducted by low-voltage scanning electron microscopy. ACTA ACUST UNITED AC 2017. [DOI: 10.1134/s1990519x17030051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
16
|
Sviben D, Forcic D, Ivancic-Jelecki J, Halassy B, Brgles M. Recovery of infective virus particles in ion-exchange and hydrophobic interaction monolith chromatography is influenced by particle charge and total-to-infective particle ratio. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1054:10-19. [PMID: 28415019 DOI: 10.1016/j.jchromb.2017.04.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/10/2017] [Accepted: 04/06/2017] [Indexed: 10/19/2022]
Abstract
Viral particles are used in medical applications as vaccines or gene therapy vectors. In order to obtain product of high purity, potency and safety for medical use purification of virus particles is a prerequisite, and chromatography is gaining increased attention to meet this aim. Here, we report on the use of ion-exchange and hydrophobic interaction chromatography on monolithic columns for purification of mumps virus (MuV) and measles virus (MeV). Efficiency of the process was monitored by quantification of infective virus particles (by 50% cell culture infective dose assay) and total virus particles, and monitoring of their size (by Nanoparticle Tracking Analysis). Ion-exchange chromatography was shown to be inefficient for MuV and best results for MeV were obtained on QA column with recovery around 17%. Purification of MuV and MeV by hydrophobic interaction chromatography resulted in recoveries around 60%. Results showed that columns with small channels (d=1.4μm) are not suitable for MuV and MeV, although their size is below 400nm, whereas columns with large channels (6μm) showed to be efficient and recoveries independent on the flow rate up to 10mL/min. Heterogeneity of the virus suspension and its interday variability mostly regarding total-to-infective particle ratio was observed. Interestingly, a trend in recovery depending on the day of the harvest was also observed for both viruses, and it correlated with the total-to-infective particle ratio, indicating influence of the virus sample composition on the chromatography results.
Collapse
Affiliation(s)
- Dora Sviben
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Dubravko Forcic
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Jelena Ivancic-Jelecki
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Beata Halassy
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Marija Brgles
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia.
| |
Collapse
|
17
|
Mateescu B, Kowal EJK, van Balkom BWM, Bartel S, Bhattacharyya SN, Buzás EI, Buck AH, de Candia P, Chow FWN, Das S, Driedonks TAP, Fernández-Messina L, Haderk F, Hill AF, Jones JC, Van Keuren-Jensen KR, Lai CP, Lässer C, Liegro ID, Lunavat TR, Lorenowicz MJ, Maas SLN, Mäger I, Mittelbrunn M, Momma S, Mukherjee K, Nawaz M, Pegtel DM, Pfaffl MW, Schiffelers RM, Tahara H, Théry C, Tosar JP, Wauben MHM, Witwer KW, Nolte-'t Hoen ENM. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper. J Extracell Vesicles 2017; 6:1286095. [PMID: 28326170 PMCID: PMC5345583 DOI: 10.1080/20013078.2017.1286095] [Citation(s) in RCA: 525] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/25/2016] [Indexed: 02/07/2023] Open
Abstract
The release of RNA-containing extracellular vesicles (EV) into the extracellular milieu has been demonstrated in a multitude of different in vitro cell systems and in a variety of body fluids. RNA-containing EV are in the limelight for their capacity to communicate genetically encoded messages to other cells, their suitability as candidate biomarkers for diseases, and their use as therapeutic agents. Although EV-RNA has attracted enormous interest from basic researchers, clinicians, and industry, we currently have limited knowledge on which mechanisms drive and regulate RNA incorporation into EV and on how RNA-encoded messages affect signalling processes in EV-targeted cells. Moreover, EV-RNA research faces various technical challenges, such as standardisation of EV isolation methods, optimisation of methodologies to isolate and characterise minute quantities of RNA found in EV, and development of approaches to demonstrate functional transfer of EV-RNA in vivo. These topics were discussed at the 2015 EV-RNA workshop of the International Society for Extracellular Vesicles. This position paper was written by the participants of the workshop not only to give an overview of the current state of knowledge in the field, but also to clarify that our incomplete knowledge – of the nature of EV(-RNA)s and of how to effectively and reliably study them – currently prohibits the implementation of gold standards in EV-RNA research. In addition, this paper creates awareness of possibilities and limitations of currently used strategies to investigate EV-RNA and calls for caution in interpretation of the obtained data.
Collapse
Affiliation(s)
- Bogdan Mateescu
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zürich) , Zurich , Switzerland
| | - Emma J K Kowal
- Department of Biology, Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Bas W M van Balkom
- Department of Nephrology and Hypertension, UMC Utrecht , Utrecht , the Netherlands
| | - Sabine Bartel
- Experimental Asthma Research, Priority Area Asthma & Allergy, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL) , Borstel , Germany
| | - Suvendra N Bhattacharyya
- Department of Science and Technology, CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University , Budapest , Hungary
| | - Amy H Buck
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | | | - Franklin W N Chow
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | - Saumya Das
- Cardiovascular Research Institute, Massachusetts General Hospital , Boston , MA , USA
| | - Tom A P Driedonks
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| | | | - Franziska Haderk
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Medicine, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University , Bundoora , Australia
| | - Jennifer C Jones
- Molecular Immunogenetics & Vaccine Research Section, Vaccine Branch, CCR, NCI , Bethesda , MD , USA
| | | | - Charles P Lai
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu , Taiwan
| | - Cecilia Lässer
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA; Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Italia di Liegro
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), University of Palermo , Palermo , Italy
| | - Taral R Lunavat
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA; Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magdalena J Lorenowicz
- Center for Molecular Medicine, University Medical Center Utrecht & Regenerative Medicine Center , Utrecht , the Netherlands
| | - Sybren L N Maas
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School , Boston , MA , USA
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Institute of Technology, University of Tartu, Tartu, Estonia
| | - Maria Mittelbrunn
- Instituto de Investigación del Hospital 12 de Octubre , Madrid , Spain
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School , Frankfurt am Main , Germany
| | - Kamalika Mukherjee
- Department of Science and Technology, CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Muhammed Nawaz
- Department of Pathology and Forensic Medicine, Ribeirão Preto School of Medicine, University of Sao Paulo , Sao Paulo , Brazil
| | - D Michiel Pegtel
- Department of Pathology, Exosomes Research Group, VU University Medical Center , Amsterdam , the Netherlands
| | - Michael W Pfaffl
- Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich (TUM) Weihenstephan , Freising , Germany
| | - Raymond M Schiffelers
- Laboratory Clinical Chemistry & Haematology, University Medical Center Utrecht , Utrecht , the Netherlands
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Institute of Biomedical & Health Sciences, Hiroshima University , Hiroshima , Japan
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932 , Paris , France
| | - Juan Pablo Tosar
- Functional Genomics Unit, Institut Pasteur de Montevideo, Nuclear Research Center, Faculty of Science, Universidad de la República , Montevideo , Uruguay
| | - Marca H M Wauben
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology and Department of Neurology, The Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Esther N M Nolte-'t Hoen
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| |
Collapse
|
18
|
Kolegraff K, Bostik P, Ansari AA. Characterization and Role of Lentivirus-Associated Host Proteins. Exp Biol Med (Maywood) 2016; 231:252-63. [PMID: 16514170 DOI: 10.1177/153537020623100303] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Enveloped viruses obtain their envelopes during the process of budding from infected cells. During this process, however, these viruses acquire parts of the host cell membranes and host cell-derived proteins as integral parts of their mature envelopes. These host-derived components of viral envelopes may subsequently exhibit various effects on the life cycle of the virus; virus cell interactions, especially host response to virus-incorporated self-proteins; and the pathogenesis of the disease induced by these viruses. Although it was known for some time that various viruses incorporate host cell-derived proteins, the issue of the role of these proteins has received increased attention, specifically in connection with human immunodeficiency virus (HIV) infection and development of acquired immunodeficiency syndrome (AIDS) in humans. The aim of this review is to summarize our current knowledge of the analysis and role of host-derived proteins associated with enveloped viruses, with emphasis on the potential role of these proteins in the pathogenesis of AIDS. Clearly, differences in the clinical outcome of those nonhuman primates infected with simian immunodeficiency virus (SIV) that are disease resistant compared with SIV-infected species that are disease susceptible provide a unique opportunity to determine whether differences in the incorporation of distinct sets of host proteins play a role with distinct clinical outcomes.
Collapse
Affiliation(s)
- Keli Kolegraff
- Department of Pathology and Laboratory Medicine, Emory University, WMB Room 2309, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
19
|
Li H, Wang S, Kong R, Ding W, Lee FH, Parker Z, Kim E, Learn GH, Hahn P, Policicchio B, Brocca-Cofano E, Deleage C, Hao X, Chuang GY, Gorman J, Gardner M, Lewis MG, Hatziioannou T, Santra S, Apetrei C, Pandrea I, Alam SM, Liao HX, Shen X, Tomaras GD, Farzan M, Chertova E, Keele BF, Estes JD, Lifson JD, Doms RW, Montefiori DC, Haynes BF, Sodroski JG, Kwong PD, Hahn BH, Shaw GM. Envelope residue 375 substitutions in simian-human immunodeficiency viruses enhance CD4 binding and replication in rhesus macaques. Proc Natl Acad Sci U S A 2016; 113:E3413-22. [PMID: 27247400 PMCID: PMC4914158 DOI: 10.1073/pnas.1606636113] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Most simian-human immunodeficiency viruses (SHIVs) bearing envelope (Env) glycoproteins from primary HIV-1 strains fail to infect rhesus macaques (RMs). We hypothesized that inefficient Env binding to rhesus CD4 (rhCD4) limits virus entry and replication and could be enhanced by substituting naturally occurring simian immunodeficiency virus Env residues at position 375, which resides at a critical location in the CD4-binding pocket and is under strong positive evolutionary pressure across the broad spectrum of primate lentiviruses. SHIVs containing primary or transmitted/founder HIV-1 subtype A, B, C, or D Envs with genotypic variants at residue 375 were constructed and analyzed in vitro and in vivo. Bulky hydrophobic or basic amino acids substituted for serine-375 enhanced Env affinity for rhCD4, virus entry into cells bearing rhCD4, and virus replication in primary rhCD4 T cells without appreciably affecting antigenicity or antibody-mediated neutralization sensitivity. Twenty-four RMs inoculated with subtype A, B, C, or D SHIVs all became productively infected with different Env375 variants-S, M, Y, H, W, or F-that were differentially selected in different Env backbones. Notably, SHIVs replicated persistently at titers comparable to HIV-1 in humans and elicited autologous neutralizing antibody responses typical of HIV-1. Seven animals succumbed to AIDS. These findings identify Env-rhCD4 binding as a critical determinant for productive SHIV infection in RMs and validate a novel and generalizable strategy for constructing SHIVs with Env glycoproteins of interest, including those that in humans elicit broadly neutralizing antibodies or bind particular Ig germ-line B-cell receptors.
Collapse
Affiliation(s)
- Hui Li
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Shuyi Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Rui Kong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Wenge Ding
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Fang-Hua Lee
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Zahra Parker
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Eunlim Kim
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Gerald H Learn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Paul Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Ben Policicchio
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | | | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Xingpei Hao
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Matthew Gardner
- Department of Infectious Disease, Scripps Research Institute, Jupiter, FL 33458
| | | | | | - Sampa Santra
- Center of Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | - S Munir Alam
- Department of Medicine, Duke University, Durham, NC 27710
| | - Hua-Xin Liao
- Department of Medicine, Duke University, Durham, NC 27710
| | - Xiaoying Shen
- Department of Medicine, Duke University, Durham, NC 27710
| | | | - Michael Farzan
- Department of Infectious Disease, Scripps Research Institute, Jupiter, FL 33458
| | - Elena Chertova
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Robert W Doms
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | | | | | - Joseph G Sodroski
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215
| | - Peter D Kwong
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Beatrice H Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| | - George M Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
20
|
Pang Y, Song H, Cheng W. Using optical trap to measure the refractive index of a single animal virus in culture fluid with high precision. BIOMEDICAL OPTICS EXPRESS 2016; 7:1672-89. [PMID: 27231613 PMCID: PMC4871073 DOI: 10.1364/boe.7.001672] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/20/2016] [Accepted: 03/29/2016] [Indexed: 05/22/2023]
Abstract
The refractive index (RI) is a fundamental parameter of materials that can be used to distinguish and sort materials of different nature. Although the RI of a virus is required for many optics-based biosensing applications, RIs of animal viruses have never been measured. Here we have developed a technique that can measure the RI of individual viruses in aqueous media with high precision. This technique is based on optical trapping of single virions and works by relating the size and RI of a single virus to the stiffness of an optical trap. We have derived an analytic expression to quantitatively describe the optical trapping of these particles. We have validated this equation using nanoparticles of known RI, and measured the RI of individual human immunodeficiency viruses type-1, which yielded a value of 1.42 at 830 nm with less than 2% coefficient of variation. This value is much lower than the RI typically assumed for viruses, but very close to that of 2.0 M sucrose solution in water. To the best of our knowledge, this is the first report on the experimental measurement of the RI for a single animal virus in aqueous media. This technique does not require prior knowledge on the diameter of the nanoparticles, and can be applied to other viruses or nanoparticles for accurate measurement of RI that is critical for the label-free detection of these particles in various settings.
Collapse
Affiliation(s)
- Yuanjie Pang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, USA
| | - Hanna Song
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, USA
| | - Wei Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
21
|
Alexander MR, Sanders RW, Moore JP, Klasse PJ. Short Communication: Virion Aggregation by Neutralizing and Nonneutralizing Antibodies to the HIV-1 Envelope Glycoprotein. AIDS Res Hum Retroviruses 2015; 31:1160-5. [PMID: 26086186 DOI: 10.1089/aid.2015.0050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We used dynamic light scattering to detect aggregation of HIV-1 virions by antibodies (IgG) to the viral envelope glycoprotein (Env). Virions of different strains were inactivated by 2,2'-dithiodipyridine (AT-2), a procedure that abrogates infectivity but preserves the native antigenic structure of Env. Neutralizing antibodies directed to a V3-base- and glycan-dependent epitope on gp120 and to the apex of the Env trimer, as well as nonneutralizing antibodies to the epitope cluster I on the gp41-ectodomain, aggregated virions, but in markedly narrow concentration ranges. In contrast, the neutralizing antibody 2G12, which is specific for a composite glycan-dependent epitope on gp120 and functionally monovalent because of its unusual domain-swap structure, was nonaggregating. These results have potentially complex implications for vaccine development.
Collapse
Affiliation(s)
- Marina R. Alexander
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| | - Rogier W. Sanders
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| | - Per Johan Klasse
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| |
Collapse
|
22
|
Exosomes and Their Role in the Life Cycle and Pathogenesis of RNA Viruses. Viruses 2015; 7:3204-25. [PMID: 26102580 PMCID: PMC4488737 DOI: 10.3390/v7062770] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 12/21/2022] Open
Abstract
Exosomes are membrane-enclosed vesicles actively released into the extracellular space, whose content reflect the physiological/pathological state of the cells they originate from. These vesicles participate in cell-to-cell communication and transfer of biologically active proteins, lipids, and RNAs. Their role in viral infections is just beginning to be appreciated. RNA viruses are an important class of pathogens and affect millions of people worldwide. Recent studies on Human Immunodeficiency Virus (HIV), Hepatitis C Virus (HCV), human T-cell lymphotropic virus (HTLV), and Dengue Virus (DENV) have demonstrated that exosomes released from infected cells harbor and deliver many regulatory factors including viral RNA and proteins, viral and cellular miRNA, and other host functional genetic elements to neighboring cells, helping to establish productive infections and modulating cellular responses. Exosomes can either spread or limit an infection depending on the type of pathogen and target cells, and can be exploited as candidates for development of antiviral or vaccine treatments. This review summarizes recent progress made in understanding the role of exosomes in RNA virus infections with an emphasis on their potential contribution to pathogenesis.
Collapse
|
23
|
Goodson P. Questioning the HIV-AIDS Hypothesis: 30 Years of Dissent. Front Public Health 2014; 2:154. [PMID: 25695040 PMCID: PMC4172096 DOI: 10.3389/fpubh.2014.00154] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/07/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Patricia Goodson
- Department of Health & Kinesiology, Texas A&M University, College Station, TX, USA
| |
Collapse
|
24
|
Müller B, Anders M, Reinstein J. In vitro analysis of human immunodeficiency virus particle dissociation: gag proteolytic processing influences dissociation kinetics. PLoS One 2014; 9:e99504. [PMID: 24915417 PMCID: PMC4051761 DOI: 10.1371/journal.pone.0099504] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 05/15/2014] [Indexed: 11/18/2022] Open
Abstract
Human immunodeficiency virus particles undergo a step of proteolytic maturation, in which the main structural polyprotein Gag is cleaved into its mature subunits matrix (MA), capsid (CA), nucleocapsid (NC) and p6. Gag proteolytic processing is accompanied by a dramatic structural rearrangement within the virion, which is necessary for virus infectivity and has been proposed to proceed through a sequence of dissociation and reformation of the capsid lattice. Morphological maturation appears to be tightly regulated, with sequential cleavage events and two small spacer peptides within Gag playing important roles by regulating the disassembly of the immature capsid layer and formation of the mature capsid lattice. In order to measure the influence of individual Gag domains on lattice stability, we established Förster's resonance energy transfer (FRET) reporter virions and employed rapid kinetic FRET and light scatter measurements. This approach allowed us to measure dissociation properties of HIV-1 particles assembled in eukaryotic cells containing Gag proteins in different states of proteolytic processing. While the complex dissociation behavior of the particles prevented an assignment of kinetic rate constants to individual dissociation steps, our analyses revealed characteristic differences in the dissociation properties of the MA layer dependent on the presence of additional domains. The most striking effect observed here was a pronounced stabilization of the MA-CA layer mediated by the presence of the 14 amino acid long spacer peptide SP1 at the CA C-terminus, underlining the crucial role of this peptide for the resolution of the immature particle architecture.
Collapse
Affiliation(s)
- Barbara Müller
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- * E-mail:
| | - Maria Anders
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jochen Reinstein
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Heidelberg, Germany
| |
Collapse
|
25
|
Mercier SK, Donaghy H, Botting RA, Turville SG, Harman AN, Nasr N, Ji H, Kusebauch U, Mendoza L, Shteynberg D, Sandgren K, Simpson RJ, Moritz RL, Cunningham AL. The microvesicle component of HIV-1 inocula modulates dendritic cell infection and maturation and enhances adhesion to and activation of T lymphocytes. PLoS Pathog 2013; 9:e1003700. [PMID: 24204260 PMCID: PMC3798598 DOI: 10.1371/journal.ppat.1003700] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 08/26/2013] [Indexed: 01/05/2023] Open
Abstract
HIV-1 is taken up by immature monocyte derived dendritic cells (iMDDCs) into tetraspanin rich caves from which the virus can either be transferred to T lymphocytes or enter into endosomes resulting in degradation. HIV-1 binding and fusion with the DC membrane results in low level de novo infection that can also be transferred to T lymphocytes at a later stage. We have previously reported that HIV-1 can induce partial maturation of iMDDCs at both stages of trafficking. Here we show that CD45⁺ microvesicles (MV) which contaminate purified HIV-1 inocula due to similar size and density, affect DC maturation, de novo HIV-1 infection and transfer to T lymphocytes. Comparing iMDDCs infected with CD45-depleted HIV-1BaL or matched non-depleted preparations, the presence of CD45⁺ MVs was shown to enhance DC maturation and ICAM-1 (CD54) expression, which is involved in DC∶T lymphocyte interactions, while restricting HIV-1 infection of MDDCs. Furthermore, in the DC culture HIV-1 infected (p24⁺) MDDCs were more mature than bystander cells. Depletion of MVs from the HIV-1 inoculum markedly inhibited DC∶T lymphocyte clustering and the induction of alloproliferation as well as limiting HIV-1 transfer from DCs to T lymphocytes. The effects of MV depletion on these functions were reversed by the re-addition of purified MVs from activated but not non-activated SUPT1.CCR5-CL.30 or primary T cells. Analysis of the protein complement of these MVs and of these HIV-1 inocula before and after MV depletion showed that Heat Shock Proteins (HSPs) and nef were the likely DC maturation candidates. Recombinant HSP90α and β and nef all induced DC maturation and ICAM-1 expression, greater when combined. These results suggest that MVs contaminating HIV-1 released from infected T lymphocytes may be biologically important, especially in enhancing T cell activation, during uptake by DCs in vitro and in vivo, particularly as MVs have been detected in the circulation of HIV-1 infected subjects.
Collapse
Affiliation(s)
- Sarah K. Mercier
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Heather Donaghy
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- * E-mail: (HD); (ALC)
| | - Rachel A. Botting
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Stuart G. Turville
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Andrew N. Harman
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Najla Nasr
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Hong Ji
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Ulrike Kusebauch
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Luis Mendoza
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - David Shteynberg
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Kerrie Sandgren
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Richard J. Simpson
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Robert L. Moritz
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Anthony L. Cunningham
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
- * E-mail: (HD); (ALC)
| |
Collapse
|
26
|
Linde ME, Colquhoun DR, Ubaida Mohien C, Kole T, Aquino V, Cotter R, Edwards N, Hildreth JEK, Graham DR. The conserved set of host proteins incorporated into HIV-1 virions suggests a common egress pathway in multiple cell types. J Proteome Res 2013; 12:2045-54. [PMID: 23432411 DOI: 10.1021/pr300918r] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
HIV-1 incorporates a large array of host proteins into virions. Determining the host protein composition in HIV virions has technical difficulties, including copurification of microvesicles. We developed an alternative purification technique using cholesterol that differentially modulates the density of virions and microvesicles (density modification, DM) allowing for high-yield virion purification that is essential for tandem mass spectrometric and quantitative proteomic (iTRAQ) analysis. DM purified virions were analyzed using iTRAQ and validated against Optiprep (60% iodixanol) purified virions. We were able to characterize host protein incorporation in DM-purified HIV particles derived from CD4+ T-cell lines; we compared this data set to a reprocessed data set of monocyte-derived macrophages (MDM) derived HIV-1 using the same bioinformatics pipeline. Seventy-nine clustered proteins were shared between the MDM derived and T-cell derived data set. These clusters included an extensive collection of actin isoforms, HLA proteins, chaperones, and a handful of other proteins, many of which have previously been documented to interact with viral proteins. Other proteins of note were ERM proteins, the dynamin domain containing protein EH4, a phosphodiesterase, and cyclophilin A. As these proteins are incorporated in virions produced in both cell types, we hypothesize that these proteins may have direct interactions with viral proteins or may be important in the viral life cycle. Additionally, identified common set proteins are predicted to interact with >1000 related human proteins. Many of these secondary interacting proteins are reported to be incorporated into virions, including ERM proteins and adhesion molecules. Thus, only a few direct interactions between host and viral proteins may dictate the host protein composition in virions. Ultimately, interaction and expression differences in host proteins between cell types may drive virion phenotypic diversity, despite conserved viral protein-host protein interactions between cell types.
Collapse
Affiliation(s)
- Michael E Linde
- Graduate Program in Immunology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Santos S, Obukhov Y, Nekhai S, Bukrinsky M, Iordanskiy S. Virus-producing cells determine the host protein profiles of HIV-1 virion cores. Retrovirology 2012; 9:65. [PMID: 22889230 PMCID: PMC3432596 DOI: 10.1186/1742-4690-9-65] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 07/15/2012] [Indexed: 11/10/2022] Open
Abstract
Background Upon HIV entry into target cells, viral cores are released and rearranged into reverse transcription complexes (RTCs), which support reverse transcription and also protect and transport viral cDNA to the site of integration. RTCs are composed of viral and cellular proteins that originate from both target and producer cells, the latter entering the target cell within the viral core. However, the proteome of HIV-1 viral cores in the context of the type of producer cells has not yet been characterized. Results We examined the proteomic profiles of the cores purified from HIV-1 NL4-3 virions assembled in Sup-T1 cells (T lymphocytes), PMA and vitamin D3 activated THP1 (model of macrophages, mMΦ), and non-activated THP1 cells (model of monocytes, mMN) and assessed potential involvement of identified proteins in the early stages of infection using gene ontology information and data from genome-wide screens on proteins important for HIV-1 replication. We identified 202 cellular proteins incorporated in the viral cores (T cells: 125, mMΦ: 110, mMN: 90) with the overlap between these sets limited to 42 proteins. The groups of RNA binding (29), DNA binding (17), cytoskeleton (15), cytoskeleton regulation (21), chaperone (18), vesicular trafficking-associated (12) and ubiquitin-proteasome pathway-associated proteins (9) were most numerous. Cores of the virions from SupT1 cells contained twice as many RNA binding proteins as cores of THP1-derived virus, whereas cores of virions from mMΦ and mMN were enriched in components of cytoskeleton and vesicular transport machinery, most probably due to differences in virion assembly pathways between these cells. Spectra of chaperones, cytoskeletal proteins and ubiquitin-proteasome pathway components were similar between viral cores from different cell types, whereas DNA-binding and especially RNA-binding proteins were highly diverse. Western blot analysis showed that within the group of overlapping proteins, the level of incorporation of some RNA binding (RHA and HELIC2) and DNA binding proteins (MCM5 and Ku80) in the viral cores from T cells was higher than in the cores from both mMΦ and mMN and did not correlate with the abundance of these proteins in virus producing cells. Conclusions Profiles of host proteins packaged in the cores of HIV-1 virions depend on the type of virus producing cell. The pool of proteins present in the cores of all virions is likely to contain factors important for viral functions. Incorporation ratio of certain RNA- and DNA-binding proteins suggests their more efficient, non-random packaging into virions in T cells than in mMΦ and mMN.
Collapse
Affiliation(s)
- Steven Santos
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, 2300 I Street NW, Ross Hall, Washington, DC 20037, USA
| | | | | | | | | |
Collapse
|
28
|
Hogue IB, Llewellyn GN, Ono A. Dynamic Association between HIV-1 Gag and Membrane Domains. Mol Biol Int 2012; 2012:979765. [PMID: 22830021 PMCID: PMC3399408 DOI: 10.1155/2012/979765] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Accepted: 06/01/2012] [Indexed: 12/18/2022] Open
Abstract
HIV-1 particle assembly is driven by the structural protein Gag. Gag binds to and multimerizes on the inner leaflet of the plasma membrane, eventually resulting in formation of spherical particles. During virus spread among T cells, Gag accumulates to the plasma membrane domain that, together with target cell membrane, forms a cell junction known as the virological synapse. While Gag association with plasma membrane microdomains has been implicated in virus assembly and cell-to-cell transmission, recent studies suggest that, rather than merely accumulating to pre-existing microdomains, Gag plays an active role in reorganizing the microdomains via its multimerization activity. In this paper, we will discuss this emerging view of Gag microdomain interactions. Relationships between Gag multimerization and microdomain association will be further discussed in the context of Gag localization to T-cell uropods and virological synapses.
Collapse
Affiliation(s)
- Ian B. Hogue
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - G. Nicholas Llewellyn
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
29
|
Coleman BM, Hanssen E, Lawson VA, Hill AF. Prion‐infected cells regulate the release of exosomes with distinct ultrastructural features. FASEB J 2012; 26:4160-73. [DOI: 10.1096/fj.11-202077] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Bradley M. Coleman
- Department of Biochemistry and Molecular BiologyThe University of MelbourneParkvilleVictoriaAustralia
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Eric Hanssen
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Electron Microscopy UnitThe University of MelbourneParkvilleVictoriaAustralia
| | - Victoria A. Lawson
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
- The Mental Health Research InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Andrew F. Hill
- Department of Biochemistry and Molecular BiologyThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
- The Mental Health Research InstituteThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
30
|
Ducloux C, Mougel M, Goldschmidt V, Didierlaurent L, Marquet R, Isel C. A pyrophosphatase activity associated with purified HIV-1 particles. Biochimie 2012; 94:2498-507. [PMID: 22766015 DOI: 10.1016/j.biochi.2012.06.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/22/2012] [Indexed: 01/17/2023]
Abstract
Treatment of HIV-1 with nucleoside reverse transcription inhibitors leads to the emergence of resistance mutations in the reverse transcriptase (RT) gene. Resistance to 3'-azido-3'-deoxythymidine (AZT) and to a lesser extent to 2'-3'-didehydro-2'-3'-dideoxythymidine is mediated by phosphorolytic excision of the chain terminator. Wild-type RT excises AZT by pyrophosphorolysis, while thymidine-associated resistance mutations in RT (TAMs) favour ATP as the donor substrate. However, in vitro, resistant RT still uses pyrophosphate more efficiently than ATP. We performed in vitro (-) strong-stop DNA synthesis experiments, with wild-type and AZT-resistant HIV-1 RTs, in the presence of physiologically relevant pyrophosphate and/or ATP concentrations and found that in the presence of pyrophosphate, ATP and AZTTP, TAMs do not enhance in vitro (-) strong-stop DNA synthesis. We hypothesized that utilisation of ATP in vivo is driven by intrinsic low pyrophosphate concentrations within the reverse transcription complex, which could be explained by the packaging of a cellular pyrophosphatase. We showed that over-expressed flagged-pyrophosphatase was associated with HIV-1 viral-like particles. In addition, we demonstrated that when HIV-1 particles were purified in order to avoid cellular microvesicle contamination, a pyrophosphatase activity was specifically associated to them. The presence of a pyrophosphatase activity in close proximity to the reverse transcription complex is most likely advantageous to the virus, even in the absence of any drug pressure.
Collapse
Affiliation(s)
- Céline Ducloux
- Architecture et Réactivité de l'ARN, Université de Strasbourg, CNRS, IBMC, 15 Rue René Descartes, 67084 Strasbourg, France.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Ubiquitin is important for the budding of many retroviruses and other enveloped viruses, but the precise role of ubiquitin in virus budding remains unclear. Here, we characterized the ubiquitination of the matrix (M) protein of a paramyxovirus, parainfluenza virus 5 (PIV5). The PIV5 M protein (but not the PIV5 nucleocapsid protein) was found to be targeted for monoubiquitination in transfected mammalian cells. Major sites of ubiquitin attachment identified by mass spectrometry analysis were lysine residues at amino acid positions 79/80, 130, and 247. The cumulative mutation of lysine residues 79, 80, and 130 to arginines led to an altered pattern of M protein ubiquitination and impaired viruslike particle (VLP) production. However, the cumulative mutation of lysine residues 79, 80, 130, and 247 to arginines restored M protein ubiquitination and VLP production, suggesting that ubiquitin is attached to alternative sites on the M protein when the primary ones have been removed. Additional lysine residues were targeted for mutagenesis based on the UbiPred algorithm. An M protein with seven lysine residues changed to arginines exhibited altered ubiquitination and poor VLP production. A recombinant virus encoding an M protein with seven lysines mutated was generated, and this virus exhibited a 6-fold-reduced maximum titer, with the defect being attributed mainly to the budding of noninfectious particles. The recombinant virus was assembly deficient, as judged by the redistribution of viral M and hemagglutinin-neuraminidase proteins in infected cells. Similar assembly defects were observed for the wild-type (wt) virus after treatment with a proteasome inhibitor. Collectively, these findings suggest that the monoubiquitination of the PIV5 M protein is important for proper virus assembly and for the budding of infectious particles.
Collapse
|
32
|
Abstract
Cells secrete various membrane-enclosed microvesicles from their cell surface (shedding microvesicles) and from internal, endosome-derived membranes (exosomes). Intriguingly, these vesicles have many characteristics in common with enveloped viruses, including biophysical properties, biogenesis, and uptake by cells. Recent discoveries describing the microvesicle-mediated intercellular transfer of functional cellular proteins, RNAs, and mRNAs have revealed additional similarities between viruses and cellular microvesicles. Apparent differences include the complexity of viral entry, temporally regulated viral expression, and self-replication proceeding to infection of new cells. Interestingly, many virally infected cells secrete microvesicles that differ in content from their virion counterparts but may contain various viral proteins and RNAs. For the most part, these particles have not been analyzed for their content or functions during viral infection. However, early studies of microvesicles (L-particles) secreted from herpes simplex virus-infected cells provided the first evidence of microvesicle-mediated intercellular communication. In the case of Epstein-Barr virus, recent evidence suggests that this tumorigenic herpesvirus also utilizes exosomes as a mechanism of cell-to-cell communication through the transfer of signaling competent proteins and functional microRNAs to uninfected cells. This review focuses on aspects of the biology of microvesicles with an emphasis on their potential contributions to viral infection and pathogenesis.
Collapse
|
33
|
Ammersbach M, Bienzle D. Methods for assessing feline immunodeficiency virus infection, infectivity and purification. Vet Immunol Immunopathol 2011; 143:202-14. [DOI: 10.1016/j.vetimm.2011.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Giroud C, Chazal N, Briant L. Cellular kinases incorporated into HIV-1 particles: passive or active passengers? Retrovirology 2011; 8:71. [PMID: 21888651 PMCID: PMC3182982 DOI: 10.1186/1742-4690-8-71] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 09/02/2011] [Indexed: 11/10/2022] Open
Abstract
Phosphorylation is one of the major mechanisms by which the activities of protein factors can be regulated. Such regulation impacts multiple key-functions of mammalian cells, including signal transduction, nucleo-cytoplasmic shuttling, macromolecular complexes assembly, DNA binding and regulation of enzymatic activities to name a few. To ensure their capacities to replicate and propagate efficiently in their hosts, viruses may rely on the phosphorylation of viral proteins to assist diverse steps of their life cycle. It has been known for several decades that particles from diverse virus families contain some protein kinase activity. While large DNA viruses generally encode for viral kinases, RNA viruses and more precisely retroviruses have acquired the capacity to hijack the signaling machinery of the host cell and to embark cellular kinases when budding. Such property was demonstrated for HIV-1 more than a decade ago. This review summarizes the knowledge acquired in the field of HIV-1-associated kinases and discusses their possible function in the retroviral life cycle.
Collapse
Affiliation(s)
- Charline Giroud
- Centre d'Études d'Agents Pathogènes et Biotechnologies pour la Santé, UMR5236 CNRS - Université Montpellier 1-Montpellier 2, Montpellier, France
| | | | | |
Collapse
|
35
|
Gag induces the coalescence of clustered lipid rafts and tetraspanin-enriched microdomains at HIV-1 assembly sites on the plasma membrane. J Virol 2011; 85:9749-66. [PMID: 21813604 DOI: 10.1128/jvi.00743-11] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The HIV-1 structural protein Gag associates with two types of plasma membrane microdomains, lipid rafts and tetraspanin-enriched microdomains (TEMs), both of which have been proposed to be platforms for HIV-1 assembly. However, a variety of studies have demonstrated that lipid rafts and TEMs are distinct microdomains in the absence of HIV-1 infection. To measure the impact of Gag on microdomain behaviors, we took advantage of two assays: an antibody-mediated copatching assay and a Förster resonance energy transfer (FRET) assay that measures the clustering of microdomain markers in live cells without antibody-mediated patching. We found that lipid rafts and TEMs copatched and clustered to a greater extent in the presence of membrane-bound Gag in both assays, suggesting that Gag induces the coalescence of lipid rafts and TEMs. Substitutions in membrane binding motifs of Gag revealed that, while Gag membrane binding is necessary to induce coalescence of lipid rafts and TEMs, either acylation of Gag or binding of phosphatidylinositol-(4,5)-bisphosphate is sufficient. Finally, a Gag derivative that is defective in inducing membrane curvature appeared less able to induce lipid raft and TEM coalescence. A higher-resolution analysis of assembly sites by correlative fluorescence and scanning electron microscopy showed that coalescence of clustered lipid rafts and TEMs occurs predominantly at completed cell surface virus-like particles, whereas a transmembrane raft marker protein appeared to associate with punctate Gag fluorescence even in the absence of cell surface particles. Together, these results suggest that different membrane microdomain components are recruited in a stepwise manner during assembly.
Collapse
|
36
|
Singethan K, Schneider-Schaulies J. Tetraspanins: Small transmembrane proteins with big impact on membrane microdomain structures. Commun Integr Biol 2011; 1:11-3. [PMID: 19704780 DOI: 10.4161/cib.1.1.6406] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 06/10/2008] [Indexed: 01/15/2023] Open
Abstract
Members of the tetraspanin family of transmembrane proteins including CD9, CD37, CD53, CD63, CD81, CD82, CD151, etc., contribute to the structural organization of the plasma membrane by forming microdomain structures, influencing cell fusion and regulating cell motility. Interestingly, K41, a CD9-specific monoclonal antibody (mAb), inhibits the release of human immunodeficiency virus (HIV-1), and the canine distemper virus (CDV)-, but not measles virus (MV)-induced cell-cell fusion. This mAb, which recognizes a conformational epitope on the large extracellular loop (LEL) of CD9, induced rapid relocation and clustering of CD9 in net-like structures at cell-cell contact areas.1 High-resolution analyses revealed that CD9 clustering is accompanied by the formation of microvilli that protrude from either side of adjacent cell surfaces, thus forming structures like microvilli zippers. While the cellular CD9-associated proteins beta1-integrin and EWI-F were co-clustered with CD9 at cell-cell interfaces, viral proteins in infected cells were differentially affected. MV envelope proteins were detected within, whereas CDV proteins were excluded from CD9 clusters, and thus, the tetraspanin CD9 can regulate cell-cell fusion by controlling the access of the viral fusion machinery to cell contact areas.
Collapse
Affiliation(s)
- Katrin Singethan
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg, Germany
| | | |
Collapse
|
37
|
Raymond A, Campbell-Sims T, Khan M, Lang M, Huang M, Bond V, Powell M. HIV Type 1 Nef is released from infected cells in CD45(+) microvesicles and is present in the plasma of HIV-infected individuals. AIDS Res Hum Retroviruses 2011; 27:167-78. [PMID: 20964480 DOI: 10.1089/aid.2009.0170] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
HIV-1 Nef has been demonstrated to be integral for viral persistence, infectivity, and the acceleration of disease pathogenesis (AIDS) in humans. Nef has also been detected in the plasma of HIV-infected individuals and is released from infected cells. The form in which Nef is released from infected cells is unknown. However, Nef is a myristoylated protein and has been shown to interact with the intracellular vesicular trafficking network. Here we show that Nef is released in CD45-containing microvesicles. This microvesicular Nef (mvNef) is detected in the plasma of HIV-infected individuals at relatively high concentrations (10 ng/ml). It is also present in tissue culture supernatants of Jurkat cells infected with HIV(MN). Interestingly, plasma mvNef levels in HIV(+) patients did not significantly correlate with viral load or CD4 count. Microvesicular Nef levels persisted in the plasma of HIV-infected individuals despite the use of antiretroviral therapy, even in individuals with undetectable viral loads. Using cell lines, we found Nef microvesicles induce apoptosis in Jurkat T-lymphocytes but had no observed effect on the U937 monocytic cell line. Given the large amount of mvNef present in the plasma of HIV-infected individuals, the apoptotic effect of mvNef on T cells, and the observed functions of extracellular soluble Nef in vitro, it seems likely that in vivo mvNef may play a significant role in the pathogenesis of AIDS.
Collapse
Affiliation(s)
| | | | - M. Khan
- Morehouse School of Medicine, Atlanta, Georgia
| | - M. Lang
- Morehouse School of Medicine, Atlanta, Georgia
| | - M.B. Huang
- Morehouse School of Medicine, Atlanta, Georgia
| | - V.C. Bond
- Morehouse School of Medicine, Atlanta, Georgia
| | - M.D. Powell
- Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
38
|
Segura MM, Kamen AA, Garnier A. Overview of current scalable methods for purification of viral vectors. Methods Mol Biol 2011; 737:89-116. [PMID: 21590394 DOI: 10.1007/978-1-61779-095-9_4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As a result of the growing interest in the use of viruses for gene therapy and vaccines, many virus-based products are being developed. The manufacturing of viruses poses new challenges for process developers and regulating authorities that need to be addressed to ensure quality, efficacy, and safety of the final product. The design of suitable purification strategies will depend on a multitude of variables including the vector production system and the nature of the virus. In this chapter, we provide an overview of the most commonly used purification methods for viral gene therapy vectors. Current chromatography options available for large-scale purification of γ-retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes simplex virus, baculovirus, and poxvirus vectors are presented.
Collapse
Affiliation(s)
- María Mercedes Segura
- Department of Biochemistry and Molecular Biology, Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | | | | |
Collapse
|
39
|
Park IW, He JJ. HIV-1 is budded from CD4+ T lymphocytes independently of exosomes. Virol J 2010; 7:234. [PMID: 20846372 PMCID: PMC2945958 DOI: 10.1186/1743-422x-7-234] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 09/16/2010] [Indexed: 01/29/2023] Open
Abstract
The convergence of HIV-1 budding and exosome biogenesis at late endosomal compartments called multivesicular bodies has fueled the debate on whether HIV-1 is budded from its target cells and transmitted in the form of exosomes. The point of contention appears to primarily derive from the types of target cells in question and lack of a well-defined protocol to separate exosomes from HIV-1. In this study, we adapted and established a simplified protocol to define the relationship between HIV-1 production and exosome biogenesis. Importantly, we took advantage of the newly established protocol to unequivocally show that HIV-1 was produced from CD4+ T lymphocytes Jurkat cells independently of exosomes. Thus, this study not only presents a simplified way to obtain highly purified HIV-1 virions for identification of host proteins packaged into virions, but also provides a technical platform that can be employed to define the relationship between exosome biogenesis and budding of HIV-1 or other viruses and its contributions to viral pathogenesis.
Collapse
Affiliation(s)
- In-Woo Park
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
40
|
Dobrowsky TM, Daniels BR, Siliciano RF, Sun SX, Wirtz D. Organization of cellular receptors into a nanoscale junction during HIV-1 adhesion. PLoS Comput Biol 2010; 6:e1000855. [PMID: 20657663 PMCID: PMC2904768 DOI: 10.1371/journal.pcbi.1000855] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 06/04/2010] [Indexed: 12/11/2022] Open
Abstract
The fusion of the human immunodeficiency virus type 1 (HIV-1) with its host cell is the target for new antiretroviral therapies. Viral particles interact with the flexible plasma membrane via viral surface protein gp120 which binds its primary cellular receptor CD4 and subsequently the coreceptor CCR5. However, whether and how these receptors become organized at the adhesive junction between cell and virion are unknown. Here, stochastic modeling predicts that, regarding binding to gp120, cellular receptors CD4 and CCR5 form an organized, ring-like, nanoscale structure beneath the virion, which locally deforms the plasma membrane. This organized adhesive junction between cell and virion, which we name the viral junction, is reminiscent of the well-characterized immunological synapse, albeit at much smaller length scales. The formation of an organized viral junction under multiple physiopathologically relevant conditions may represent a novel intermediate step in productive infection. The entry of human immunodeficiency virus (HIV) into cells is the target for new therapies preventing HIV infection. While intermediate steps of viral entry have been characterized, the progression between these steps and how they result in productive infection are not well understood. By using stochastic modeling, we examine the initial interaction of a single viral particle with a flexible plasma membrane populated with viral receptors. The model predicts the formation of an organized receptor ultrastructure beneath the viral particle, which we name viral junction and which may contribute to productive viral infection. The organization of the viral junction depends on receptor density, CD4 bond stability, membrane mechanical flexibility, as well as viral protein organization and density.
Collapse
Affiliation(s)
- Terrence M. Dobrowsky
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Brian R. Daniels
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Robert F. Siliciano
- Howard Hughes Medical Institute and Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Sean X. Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences in Oncology Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences in Oncology Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abstract
Tetraspanins are small integral membrane proteins that are known to control a variety of cellular processes, including signaling, migration and cell-cell fusion. Research over the past few years established that they are also regulators of various steps in the HIV-1 replication cycle, but the mechanisms through which these proteins either enhance or repress virus spread remain largely unknown.
Collapse
Affiliation(s)
- Markus Thali
- Department of Microbiology and Molecular Genetics, College of Medicine and CALS, University of Vermont, 318 Stafford Hall, 95 Carrigan Drive, Burlington, VT 05405-0084, USA.
| |
Collapse
|
42
|
Leaman DP, Kinkead H, Zwick MB. In-solution virus capture assay helps deconstruct heterogeneous antibody recognition of human immunodeficiency virus type 1. J Virol 2010; 84:3382-95. [PMID: 20089658 PMCID: PMC2838137 DOI: 10.1128/jvi.02363-09] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 01/06/2010] [Indexed: 01/27/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) on whole virions is heterogeneous, so molecular analysis of Env with monoclonal antibodies (MAbs) is challenging. Virus capture assays (VCAs) involving immobilized MAbs are typically used, but these assays suffer from immobilization artifacts and do not provide binding constants. Furthermore, we show here that certain HIV-1 neutralizing MAbs, including 2G12, 4E10, 2F5, Z13e1, and D5, will capture virion particles completely devoid of Env. We modified the VCA such that MAbs and virions are incubated in solution, and unbound MAbs are removed prior to the capture step. This modification nearly eliminated evidence of Env-independent binding by MAbs to virions and allowed determination of apparent affinity constants in solution. Three important qualitative observations were further revealed. First, neutralizing MAbs 2F5, 4E10, and Z13e1 against the membrane-proximal external region (MPER) of HIV-1 gp41 were found to capture virions efficiently only if a significant amount of uncleaved gp160 or synthetic MPER peptide was present. Second, we show how non-native forms of Env vary by Env genotype and that Env from HIV-1(JR-FL) is more homogeneously trimeric than that from HIV-1(JR-CSF). Third, we determined that Env containing all or parts of gp41, including uncleaved gp160, binds spontaneously to free virions. This exogenous Env is an indiscriminate molecular "bridge" between Env-specific Ab and virions and can affect VCA analyses, particularly using pseudotyped virions. Heterogeneity in Env from endogenous and exogenous sources might also subvert humoral immunity to HIV-1, so in-solution VCAs may help to dissect this heterogeneity for vaccine design purposes.
Collapse
Affiliation(s)
- Daniel P. Leaman
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
| | - Heather Kinkead
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
| | - Michael B. Zwick
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037
| |
Collapse
|
43
|
Role of HIV membrane in neutralization by two broadly neutralizing antibodies. Proc Natl Acad Sci U S A 2009; 106:20234-9. [PMID: 19906992 DOI: 10.1073/pnas.0908713106] [Citation(s) in RCA: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Induction of effective antibody responses against HIV-1 infection remains an elusive goal for vaccine development. Progress may require in-depth understanding of the molecular mechanisms of neutralization by monoclonal antibodies. We have analyzed the molecular actions of two rare, broadly neutralizing, human monoclonal antibodies, 4E10 and 2F5, which target the transiently exposed epitopes in the membrane proximal external region (MPER) of HIV-1 gp41 envelope during viral entry. Both have long CDR H3 loops with a hydrophobic surface facing away from the peptide epitope. We find that the hydrophobic residues of 4E10 mediate a reversible attachment to the viral membrane and that they are essential for neutralization, but not for interaction with gp41. We propose that these antibodies associate with the viral membrane in a required first step and are thereby poised to capture the transient gp41 fusion intermediate. These results bear directly on strategies for rational design of HIV-1 envelope immunogens.
Collapse
|
44
|
Tumultuous relationship between the human immunodeficiency virus type 1 viral infectivity factor (Vif) and the human APOBEC-3G and APOBEC-3F restriction factors. Microbiol Mol Biol Rev 2009; 73:211-32. [PMID: 19487726 DOI: 10.1128/mmbr.00040-08] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The viral infectivity factor (Vif) is dispensable for human immunodeficiency virus type 1 (HIV-1) replication in so-called permissive cells but is required for replication in nonpermissive cell lines and for pathogenesis. Virions produced in the absence of Vif have an aberrant morphology and an unstable core and are unable to complete reverse transcription. Recent studies demonstrated that human APOBEC-3G (hA3G) and APOBEC-3F (hA3F), which are selectively expressed in nonpermissive cells, possess strong anti-HIV-1 activity and are sufficient to confer a nonpermissive phenotype. Vif induces the degradation of hA3G and hA3F, suggesting that its main function is to counteract these cellular factors. Most studies focused on the hypermutation induced by the cytidine deaminase activity of hA3G and hA3F and on their Vif-induced degradation by the proteasome. However, recent studies suggested that several mechanisms are involved both in the antiviral activity of hA3G and hA3F and in the way Vif counteracts these antiviral factors. Attempts to reconcile the studies involving Vif in virus assembly and stability with these recent findings suggest that hA3G and hA3F partially exert their antiviral activity independently of their catalytic activity by destabilizing the viral core and the reverse transcription complex, possibly by interfering with the assembly and/or maturation of the viral particles. Vif could then counteract hA3G and hA3F by excluding them from the viral assembly intermediates through competition for the viral genomic RNA, by regulating the proteolytic processing of Pr55(Gag), by enhancing the efficiency of the reverse transcription process, and by inhibiting the enzymatic activities of hA3G and hA3F.
Collapse
|
45
|
Grigorov B, Attuil-Audenis V, Perugi F, Nedelec M, Watson S, Pique C, Darlix JL, Conjeaud H, Muriaux D. A role for CD81 on the late steps of HIV-1 replication in a chronically infected T cell line. Retrovirology 2009; 6:28. [PMID: 19284574 PMCID: PMC2657109 DOI: 10.1186/1742-4690-6-28] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 03/11/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND HIV-1 uses cellular co-factors for virion formation and release. The virus is able to incorporate into the viral particles host cellular proteins, such as tetraspanins which could serve to facilitate HIV-1 egress. Here, we investigated the implication of several tetraspanins on HIV-1 formation and release in chronically infected T-lymphoblastic cells, a model that permits the study of the late steps of HIV-1 replication. RESULTS Our data revealed that HIV-1 Gag and Env structural proteins co-localized with tetraspanins in the form of clusters. Co-immunoprecipitation experiments showed that Gag proteins interact, directly or indirectly, with CD81, and less with CD82, in tetraspanin-enriched microdomains composed of CD81/CD82/CD63. In addition, when HIV-1 producing cells were treated with anti-CD81 antibodies, or upon CD81 silencing by RNA interference, HIV-1 release was significantly impaired, and its infectivity was modulated. Finally, CD81 downregulation resulted in Gag redistribution at the cell surface. CONCLUSION Our findings not only extend the notion that HIV-1 assembly can occur on tetraspanin-enriched microdomains in T cells, but also highlight a critical role for the tetraspanin CD81 on the late steps of HIV replication.
Collapse
Affiliation(s)
- Boyan Grigorov
- LaboRetro, Unité de Virologie Humaine INSERM U758, Ecole Normale Supérieure de Lyon, Lyon, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
HIV-1 and microvesicles from T cells share a common glycome, arguing for a common origin. Nat Chem Biol 2009; 5:244-50. [PMID: 19234452 PMCID: PMC2713040 DOI: 10.1038/nchembio.151] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Accepted: 02/03/2009] [Indexed: 01/17/2023]
Abstract
HIV-1 is a master at deceiving the immune system, usurping host biosynthetic machinery. Although HIV-1 is coated with host-derived glycoproteins only glycosylation of viral gp120 has been described. Herein we utilize lectin microarray technology to analyze the glycome of intact HIV-1 virions. We show that the glycan coat of human T-cell line-derived HIV-1 matches that of native immunomodulatory microvesicles. The carbohydrate composition of both virus and microvesicles is cell-line dependent, suggesting a mechanism to rapidly camouflage the virus within the host. In addition, binding of both virus and microvesicles to antiviral lectins is enriched over the host cell, raising concern about targeting these glycans for therapeutics. This work also sheds light on the binding of HIV-1 to galectin-1, an important human immune lectin. Overall, our work strongly supports the theory that HIV-1 co-opts the exocytic pathway of microvesicles, potentially explaining why eliciting a protective antiviral immune response is difficult.
Collapse
|
47
|
Abstract
VP40, the major matrix protein of Marburg virus, is the main driving force for viral budding. Additionally, cellular factors are likely to play an important role in the release of progeny virus. In the present study, we characterized the influence of the vacuolar protein sorting (VPS) pathway on the release of virus-like particles (VLPs), which are induced by Marburg virus VP40. In the supernatants of HEK 293 cells expressing VP40, different populations of VLPs with either a vesicular or a filamentous morphology were detected. While the filaments were almost completely composed of VP40, the vesicular particles additionally contained considerable amounts of cellular proteins. In contrast to that in the vesicles, the VP40 in the filaments was regularly organized, probably inducing the elimination of cellular proteins from the released VLPs. Vesicular particles were observed in the supernatants of cells even in the absence of VP40. Mutation of the late-domain motif in VP40 resulted in reduced release of filamentous particles, and likewise, inhibition of the VPS pathway by expression of a dominant-negative (DN) form of VPS4 inhibited the release of filamentous particles. In contrast, the release of vesicular particles did not respond significantly to the expression of DN VPS4. Like the budding of VLPs, the budding of Marburg virus particles was partially inhibited by the expression of DN VPS4. While the release of VLPs from VP40-expressing cells is a valuable tool with which to investigate the budding of Marburg virus particles, it is important to separate filamentous VLPs from vesicular particles, which contain many cellular proteins and use a different budding mechanism.
Collapse
|
48
|
Yang W, Hood BL, Chadwick SL, Liu S, Watkins SC, Luo G, Conrads TP, Wang T. Fatty acid synthase is up-regulated during hepatitis C virus infection and regulates hepatitis C virus entry and production. Hepatology 2008; 48:1396-403. [PMID: 18830996 PMCID: PMC2614928 DOI: 10.1002/hep.22508] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Hepatitis C virus (HCV) is a major human pathogen that causes serious illness, including acute and chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Using a mass spectrometry-based proteomics approach, we have identified 175 proteins from a cell culture supernatant fraction containing the HCV genotype 2a (JFH1) virus, among which fatty acid synthase (FASN), the multifunctional enzyme catalyzing the de novo synthesis of fatty acids, was confirmed to be highly enriched. Subsequent studies showed that FASN expression increased in the human hepatoma cell line, Huh7, or its derivative, upon HCV infection. Blocking FASN activity by C75, a pharmacological FASN inhibitor, led to decreased HCV production. Reduction of FASN by RNA interference suppressed viral replication in both replicon and infection systems. Remarkably, FASN appeared to be selectively required for the expression of claudin-1, a tight junction protein that was recently identified as an entry coreceptor for HCV, but not for the expression of another HCV coreceptor, CD81. The decrease in Claudin-1 expression resulting from FASN inhibition was accompanied by a decrease in transepithelial electric resistance of Huh7 cells, implying a reduction in the relative tightness of the cell monolayer. Consequently, the entry of human immunodeficiency virus-HCV pseudotypes was significantly inhibited in C75-treated Huh7 cells. CONCLUSION As far as we know, this is the first line of evidence that demonstrates that HCV infection directly induces FASN expression, and thus suggests a possible mechanism by which HCV infection alters the cellular lipid profile and causes diseases such as steatosis.
Collapse
Affiliation(s)
- Wei Yang
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261,*Correspondence to Dr. Tianyi Wang or Wei Yang, Department of Infectious Diseases and Microbiology, Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15261, USA. Fax: 412-383-8926, or
| | - Brian L. Hood
- Clinical Proteomics Facility, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Sara L. Chadwick
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Shufeng Liu
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Simon C. Watkins
- Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Guangxiang Luo
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA
| | - Thomas P. Conrads
- Clinical Proteomics Facility, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Tianyi Wang
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261,*Correspondence to Dr. Tianyi Wang or Wei Yang, Department of Infectious Diseases and Microbiology, Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15261, USA. Fax: 412-383-8926, or
| |
Collapse
|
49
|
Abstract
It has been known for some time that HIV-1 virions contain cellular proteins in addition to proteins encoded by the viral genome. Recent studies have vastly increased the number of host proteins detected in HIV-1. This review summarises the current findings on several cellular proteins present in these virions, including some functional studies on their potential roles in the viral replication cycle and pathogenesis. Because retroviruses require extensive assistance from host proteins and pathways, the data from biochemical characterisations of HIV-1 serve as an important starting point for understanding the role of cellular proteins that act in or influence the biology of HIV-1. Additionally, a better understanding of the interactions between cellular proteins and viral components might provide more targets for anti-HIV therapeutic intervention and provide for a better understanding of how HIV-1 alters the immune system. The extensive study of HIV-1 has already brought new insights to the fields of immunology and vaccine science. In the same way, knowledge of viral--cellular protein interactions might assist our understanding of important cellular pathways.
Collapse
Affiliation(s)
- David E Ott
- AIDS Vaccine Program, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA.
| |
Collapse
|
50
|
Cantin R, Diou J, Bélanger D, Tremblay AM, Gilbert C. Discrimination between exosomes and HIV-1: purification of both vesicles from cell-free supernatants. J Immunol Methods 2008; 338:21-30. [PMID: 18675270 DOI: 10.1016/j.jim.2008.07.007] [Citation(s) in RCA: 236] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 06/02/2008] [Accepted: 07/01/2008] [Indexed: 01/02/2023]
Abstract
Although enveloped retroviruses bud from the cell surface of T lymphocytes, they use the endocytic pathway and the internal membrane of multivesicular bodies for their assembly and release from macrophages and dendritic cells (DCs). Exosomes, physiological nanoparticles produced by hematopoietic cells, egress from this same pathway and are similar to retroviruses in terms of size, density, the molecules they incorporate and their ability to activate immune cells. Retroviruses are therefore likely to contaminate in vitro preparations of exosomes and vice versa and sucrose gradients are inefficient at separating them. However, we have found that their sedimentation velocities in an iodixanol (Optiprep) velocity gradient are sufficiently different to allow separation and purification of both vesicles. Using acetylcholinesterase as an exosome marker, we demonstrate that Optiprep velocity gradients are very efficient in separating exosomes from HIV-1 particles produced on 293T cells, primary CD4(+) T cells, macrophages or DCs, with exosomes collecting at 8.4-12% iodixanol and HIV-1 at 15.6%. We also show that immunodepletion with an anti-acetylcholinesterase antibody rapidly produces highly purified preparations of HIV-1 or exosomes. These findings have applications in fundamental research on exosomes and/or AIDS, as well as in clinical applications where exosomes are involved, more specifically in tumour therapy or in gene therapy using exosomes generated from DCs genetically modified by transfection with virus.
Collapse
Affiliation(s)
- Réjean Cantin
- Centre de recherche en infectiologie, Faculty of Medicine, Laval University, Québec, Canada
| | | | | | | | | |
Collapse
|