1
|
Bongers R, Rochus CM, Houlahan K, Lynch C, Oliveira GA, Rojas de Oliveira H, van Staaveren N, Kelton DF, Miglior F, Schenkel FS, Baes CF. Estimation of genetic parameters and genome-wide association study for enzootic bovine leukosis resistance in Canadian Holstein cattle. J Dairy Sci 2025; 108:611-622. [PMID: 39343214 DOI: 10.3168/jds.2024-25196] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024]
Abstract
Bovine leukemia virus (BLV) is the causative agent of enzootic bovine leukosis (hereafter referred to as leukosis), frequently observed in North American dairy herds. Infection with BLV can lead to persistent lymphocytosis and tumors and is associated with decreased production, immunity. and fertility. With no available treatment or vaccine, reducing the prevalence of leukosis through management and culling has not yet been successful. Genetic selection could contribute to permanent improvement in dairy cattle resistance to leukosis. This study aimed to examine the prevalence and impact of leukosis in Canada and to assess the potential for including leukosis resistance in Canadian national genetic evaluations by characterizing the genetic architecture of leukosis resistance using pedigree and genomic information. A total of 117,349 milk ELISA test records on 96,779 Holstein cows from 950 Canadian herds taken between 2007 and 2021 were provided by Lactanet Canada (Guelph, ON, Canada). Each cow was classified as test-positive for leukosis or test-negative for leukosis. Leukosis was present in ∼77% of herds tested; within those herds, an average of 39% of cows tested were test-positive for leukosis. Heritabilities of 0.10 (SE = 0.001) and 0.07 (SE <0.001) were estimated for leukosis resistance using a linear animal model and BLUP or single-step GBLUP methodology, respectively. Breeding value correlations were estimated between leukosis resistance and economically important and phenotypically relevant traits. Most correlations between leukosis resistance and traits already included in Canadian genetic evaluations were favorable, with the exception of SCS. The candidate genes for leukosis resistance identified using a genome-wide association study were on chromosome 23, with some being part of the major histocompatibility complex. This study showed that genetic evaluation for leukosis resistance is possible, and could be considered for inclusion in Canadian national selection indices.
Collapse
Affiliation(s)
- Renee Bongers
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Christina M Rochus
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Kerry Houlahan
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Colin Lynch
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Gerson A Oliveira
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Hinayah Rojas de Oliveira
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Nienke van Staaveren
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - David F Kelton
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Filippo Miglior
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada; Lactanet Canada, Guelph, ON N1K 1E5, Canada
| | - Flavio S Schenkel
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Christine F Baes
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada; Institute of Genetics, Department of Clinical Research and Veterinary Public Health, University of Bern, Bern 3001, Switzerland.
| |
Collapse
|
2
|
Serrano T, Casartelli N, Ghasemi F, Wioland H, Cuvelier F, Salles A, Moya-Nilges M, Welker L, Bernacchi S, Ruff M, Jégou A, Romet-Lemonne G, Schwartz O, Frémont S, Echard A. HIV-1 budding requires cortical actin disassembly by the oxidoreductase MICAL1. Proc Natl Acad Sci U S A 2024; 121:e2407835121. [PMID: 39556735 PMCID: PMC11621841 DOI: 10.1073/pnas.2407835121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/30/2024] [Indexed: 11/20/2024] Open
Abstract
Many enveloped viruses bud from the plasma membrane that is tightly associated with a dense and thick actin cortex. This actin network represents a significant challenge for membrane deformation and scission, and how it is remodeled during the late steps of the viral cycle is largely unknown. Using superresolution microscopy, we show that HIV-1 buds in areas of the plasma membrane with low cortical F-actin levels. We find that the cellular oxidoreductase MICAL1 locally depolymerizes actin at budding sites to promote HIV-1 budding and release. Upon MICAL1 depletion, F-actin abnormally remains at viral budding sites, incompletely budded viruses accumulate at the plasma membrane and viral release is impaired. Remarkably, normal viral release can be restored in MICAL1-depleted cells by inhibiting Arp2/3-dependent branched actin networks. Mechanistically, we find that MICAL1 directly disassembles branched-actin networks and controls the timely recruitment of the Endosomal Sorting Complexes Required for Transport scission machinery during viral budding. In addition, the MICAL1 activator Rab35 is recruited at budding sites, functions in the same pathway as MICAL1, and is also required for viral release. This work reveals a role for oxidoreduction in triggering local actin depolymerization to control HIV-1 budding, a mechanism that may be widely used by other viruses. The debranching activity of MICAL1 could be involved beyond viral budding in various other cellular functions requiring local plasma membrane deformation.
Collapse
Affiliation(s)
- Thomas Serrano
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| | - Nicoletta Casartelli
- Virology department, Virus and Immunity Lab, Institut Pasteur, Université Paris Cité, ParisF-75015, France
| | - Foad Ghasemi
- Université Paris Cité, CNRS, Institut Jacques Monod, ParisF-75013, France
| | - Hugo Wioland
- Université Paris Cité, CNRS, Institut Jacques Monod, ParisF-75013, France
| | - Frédérique Cuvelier
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| | - Audrey Salles
- Institut Pasteur, Université Paris Cité, Photonic Bio-Imaging Unit, Centre de Ressources et Recherches Technologiques (UTechS-PBI, C2RT), ParisF-75015, France
| | - Maryse Moya-Nilges
- Institut Pasteur, Université Paris Cité, Ultrastructural BioImaging, ParisF-75015, France
| | - Lisa Welker
- Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l’ARN, CNRS UPR9002, StrasbourgF-67084, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Integrated Structural Biology, CNRS UMR 7104, Inserm U 1258, University of Strasbourg, IllkirchF-67404, France
| | - Serena Bernacchi
- Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l’ARN, CNRS UPR9002, StrasbourgF-67084, France
| | - Marc Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Integrated Structural Biology, CNRS UMR 7104, Inserm U 1258, University of Strasbourg, IllkirchF-67404, France
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, ParisF-75013, France
| | | | - Olivier Schwartz
- Virology department, Virus and Immunity Lab, Institut Pasteur, Université Paris Cité, ParisF-75015, France
| | - Stéphane Frémont
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| | - Arnaud Echard
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| |
Collapse
|
3
|
Vélez-López O, Carrasquillo-Carrión K, Cantres-Rosario YM, Machín-Martínez E, Álvarez-Ríos ME, Roche-Lima A, Tosado-Rodríguez EL, Meléndez LM. Analysis of Sigma-1 Receptor Antagonist BD1047 Effect on Upregulating Proteins in HIV-1-Infected Macrophages Exposed to Cocaine Using Quantitative Proteomics. Biomedicines 2024; 12:1934. [PMID: 39335448 PMCID: PMC11428496 DOI: 10.3390/biomedicines12091934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/08/2024] [Accepted: 08/10/2024] [Indexed: 09/30/2024] Open
Abstract
HIV-1 infects monocyte-derived macrophages (MDM) that migrate into the brain and secrete virus and neurotoxic molecules, including cathepsin B (CATB), causing cognitive dysfunction. Cocaine potentiates CATB secretion and neurotoxicity in HIV-infected MDM. Pretreatment with BD1047, a sigma-1 receptor antagonist, before cocaine exposure reduces HIV-1, CATB secretion, and neuronal apoptosis. We aimed to elucidate the intracellular pathways modulated by BD1047 in HIV-infected MDM exposed to cocaine. We hypothesized that the Sig1R antagonist BD1047, prior to cocaine, significantly deregulates proteins and pathways involved in HIV-1 replication and CATB secretion that lead to neurotoxicity. MDM culture lysates from HIV-1-infected women treated with BD1047 before cocaine were compared with untreated controls using TMT quantitative proteomics, bioinformatics, Lima statistics, and pathway analyses. Results demonstrate that pretreatment with BD1047 before cocaine dysregulated eighty (80) proteins when compared with the infected cocaine group. We found fifteen (15) proteins related to HIV-1 infection, CATB, and mitochondrial function. Upregulated proteins were related to oxidative phosphorylation (SLC25A-31), mitochondria (ATP5PD), ion transport (VDAC2-3), endoplasmic reticulum transport (PHB, TMED10, CANX), and cytoskeleton remodeling (TUB1A-C, ANXA1). BD1047 treatment protects HIV-1-infected MDM exposed to cocaine by upregulating proteins that reduce mitochondrial damage, ER transport, and exocytosis associated with CATB-induced neurotoxicity.
Collapse
Affiliation(s)
- Omar Vélez-López
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
| | - Kelvin Carrasquillo-Carrión
- Integrated Informatics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00934, USA; (K.C.-C.); (A.R.-L.); (E.L.T.-R.)
| | - Yadira M. Cantres-Rosario
- Translational Proteomics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00921, USA;
| | - Eraysy Machín-Martínez
- Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00921, USA; (E.M.-M.); (M.E.Á.-R.)
| | - Manuel E. Álvarez-Ríos
- Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00921, USA; (E.M.-M.); (M.E.Á.-R.)
| | - Abiel Roche-Lima
- Integrated Informatics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00934, USA; (K.C.-C.); (A.R.-L.); (E.L.T.-R.)
| | - Eduardo L. Tosado-Rodríguez
- Integrated Informatics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00934, USA; (K.C.-C.); (A.R.-L.); (E.L.T.-R.)
| | - Loyda M. Meléndez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
- Translational Proteomics, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00921, USA;
| |
Collapse
|
4
|
Song MS, Lee DK, Lee CY, Park SC, Yang J. Host Subcellular Organelles: Targets of Viral Manipulation. Int J Mol Sci 2024; 25:1638. [PMID: 38338917 PMCID: PMC10855258 DOI: 10.3390/ijms25031638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Viruses have evolved sophisticated mechanisms to manipulate host cell processes and utilize intracellular organelles to facilitate their replication. These complex interactions between viruses and cellular organelles allow them to hijack the cellular machinery and impair homeostasis. Moreover, viral infection alters the cell membrane's structure and composition and induces vesicle formation to facilitate intracellular trafficking of viral components. However, the research focus has predominantly been on the immune response elicited by viruses, often overlooking the significant alterations that viruses induce in cellular organelles. Gaining a deeper understanding of these virus-induced cellular changes is crucial for elucidating the full life cycle of viruses and developing potent antiviral therapies. Exploring virus-induced cellular changes could substantially improve our understanding of viral infection mechanisms.
Collapse
Affiliation(s)
- Min Seok Song
- Department of Physiology and Convergence Medical Science, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Dong-Kun Lee
- Department of Physiology and Convergence Medical Science, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Chung-Young Lee
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Sang-Cheol Park
- Artificial Intelligence and Robotics Laboratory, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Jinsung Yang
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| |
Collapse
|
5
|
Dibsy R, Bremaud E, Mak J, Favard C, Muriaux D. HIV-1 diverts cortical actin for particle assembly and release. Nat Commun 2023; 14:6945. [PMID: 37907528 PMCID: PMC10618566 DOI: 10.1038/s41467-023-41940-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
Enveloped viruses assemble and bud from the host cell membranes. Any role of cortical actin in these processes have often been a source of debate. Here, we assessed if cortical actin was involved in HIV-1 assembly in infected CD4 T lymphocytes. Our results show that preventing actin branching not only increases HIV-1 particle release but also the number of individual HIV-1 Gag assembly clusters at the T cell plasma membrane. Indeed, in infected T lymphocytes and in in vitro quantitative model systems, we show that HIV-1 Gag protein prefers areas deficient in F-actin for assembling. Finally, we found that the host factor Arpin, an inhibitor of Arp2/3 branched actin, is recruited at the membrane of infected T cells and it can associate with the viral Gag protein. Altogether, our data show that, for virus assembly and particle release, HIV-1 prefers low density of cortical actin and may favor local actin debranching by subverting Arpin.
Collapse
Affiliation(s)
- Rayane Dibsy
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Erwan Bremaud
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Johnson Mak
- Institute for Glycomics, Griffith University, Brisbane, Australia
| | - Cyril Favard
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Delphine Muriaux
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France.
| |
Collapse
|
6
|
Iida N, Kawahara M, Hirota R, Shibagaki Y, Hattori S, Morikawa Y. A Proteomic Analysis of Detergent-Resistant Membranes in HIV Virological Synapse: The Involvement of Vimentin in CD4 Polarization. Viruses 2023; 15:1266. [PMID: 37376566 DOI: 10.3390/v15061266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The cell-cell contact between HIV-1-infected and uninfected cells forms a virological synapse (VS) to allow for efficient HIV-1 transmission. Not only are HIV-1 components polarized and accumulate at cell-cell interfaces, but viral receptors and lipid raft markers are also. To better understand the nature of the HIV-1 VS, detergent-resistant membrane (DRM) fractions were isolated from an infected-uninfected cell coculture and compared to those from non-coculture samples using 2D fluorescence difference gel electrophoresis. Mass spectrometry revealed that ATP-related enzymes (ATP synthase subunit and vacuolar-type proton ATPase), protein translation factors (eukaryotic initiation factor 4A and mitochondrial elongation factor Tu), protein quality-control-related factors (protein disulfide isomerase A3 and 26S protease regulatory subunit), charged multivesicular body protein 4B, and vimentin were recruited to the VS. Membrane flotation centrifugation of the DRM fractions and confocal microscopy confirmed these findings. We further explored how vimentin contributes to the HIV-1 VS and found that vimentin supports HIV-1 transmission through the recruitment of CD4 to the cell-cell interface. Since many of the molecules identified in this study have previously been suggested to be involved in HIV-1 infection, we suggest that a 2D difference gel analysis of DRM-associated proteins may reveal the molecules that play crucial roles in HIV-1 cell-cell transmission.
Collapse
Affiliation(s)
- Naoyuki Iida
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Madoka Kawahara
- Omura Satoshi Memorial Institute and Graduate School for Infection Control, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Riku Hirota
- Omura Satoshi Memorial Institute and Graduate School for Infection Control, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Yoshio Shibagaki
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Seisuke Hattori
- School of Pharmacy, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Yuko Morikawa
- Omura Satoshi Memorial Institute and Graduate School for Infection Control, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
7
|
Mascarau R, Woottum M, Fromont L, Gence R, Cantaloube-Ferrieu V, Vahlas Z, Lévêque K, Bertrand F, Beunon T, Métais A, El Costa H, Jabrane-Ferrat N, Gallois Y, Guibert N, Davignon JL, Favre G, Maridonneau-Parini I, Poincloux R, Lagane B, Bénichou S, Raynaud-Messina B, Vérollet C. Productive HIV-1 infection of tissue macrophages by fusion with infected CD4+ T cells. J Cell Biol 2023; 222:213978. [PMID: 36988579 PMCID: PMC10067447 DOI: 10.1083/jcb.202205103] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 12/05/2022] [Accepted: 02/02/2023] [Indexed: 03/30/2023] Open
Abstract
Macrophages are essential for HIV-1 pathogenesis and represent major viral reservoirs. Therefore, it is critical to understand macrophage infection, especially in tissue macrophages, which are widely infected in vivo, but poorly permissive to cell-free infection. Although cell-to-cell transmission of HIV-1 is a determinant mode of macrophage infection in vivo, how HIV-1 transfers toward macrophages remains elusive. Here, we demonstrate that fusion of infected CD4+ T lymphocytes with human macrophages leads to their efficient and productive infection. Importantly, several tissue macrophage populations undergo this heterotypic cell fusion, including synovial, placental, lung alveolar, and tonsil macrophages. We also find that this mode of infection is modulated by the macrophage polarization state. This fusion process engages a specific short-lived adhesion structure and is controlled by the CD81 tetraspanin, which activates RhoA/ROCK-dependent actomyosin contractility in macrophages. Our study provides important insights into the mechanisms underlying infection of tissue-resident macrophages, and establishment of persistent cellular reservoirs in patients.
Collapse
Affiliation(s)
- Rémi Mascarau
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
- International Research Project " MAC-TB/HIV " , Toulouse, France
| | - Marie Woottum
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris , Paris, France
| | - Léa Fromont
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
| | - Rémi Gence
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037 and Institut Universitaire du Cancer de Toulouse - Oncopôle , Toulouse, France
| | - Vincent Cantaloube-Ferrieu
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université Toulouse, Centre National de la Recherche Scientifique, Inserm , Toulouse, France
| | - Zoï Vahlas
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
- International Research Project " MAC-TB/HIV " , Toulouse, France
| | - Kevin Lévêque
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
| | - Florent Bertrand
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
| | - Thomas Beunon
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
| | - Arnaud Métais
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
| | - Hicham El Costa
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université Toulouse, Centre National de la Recherche Scientifique, Inserm , Toulouse, France
| | - Nabila Jabrane-Ferrat
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université Toulouse, Centre National de la Recherche Scientifique, Inserm , Toulouse, France
| | - Yohan Gallois
- ENT, Otoneurology and Pediatric ENT Department, University Hospital of Toulouse , Toulouse, France
| | - Nicolas Guibert
- Thoracic Endoscopy Unit, Pulmonology Department, Larrey University Hospital , Toulouse, France
| | | | - Gilles Favre
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037 and Institut Universitaire du Cancer de Toulouse - Oncopôle , Toulouse, France
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
- International Research Project " MAC-TB/HIV " , Toulouse, France
| | - Renaud Poincloux
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
- International Research Project " MAC-TB/HIV " , Toulouse, France
| | - Bernard Lagane
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université Toulouse, Centre National de la Recherche Scientifique, Inserm , Toulouse, France
| | - Serge Bénichou
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique UMR8104, Université de Paris , Paris, France
| | - Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
- International Research Project " MAC-TB/HIV " , Toulouse, France
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS) , Toulouse, France
- International Research Project " MAC-TB/HIV " , Toulouse, France
| |
Collapse
|
8
|
Río-Bergé C, Cong Y, Reggiori F. Getting on the right track: Interactions between viruses and the cytoskeletal motor proteins. Traffic 2023; 24:114-130. [PMID: 35146839 DOI: 10.1111/tra.12835] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 11/29/2022]
Abstract
The cytoskeleton is an essential component of the cell and it is involved in multiple physiological functions, including intracellular organization and transport. It is composed of three main families of proteinaceous filaments; microtubules, actin filaments and intermediate filaments and their accessory proteins. Motor proteins, which comprise the dynein, kinesin and myosin superfamilies, are a remarkable group of accessory proteins that mainly mediate the intracellular transport of cargoes along with the cytoskeleton. Like other cellular structures and pathways, viruses can exploit the cytoskeleton to promote different steps of their life cycle through associations with motor proteins. The complexity of the cytoskeleton and the differences among viruses, however, has led to a wide diversity of interactions, which in most cases remain poorly understood. Unveiling the details of these interactions is necessary not only for a better comprehension of specific infections, but may also reveal new potential drug targets to fight dreadful diseases such as rabies disease and acquired immunodeficiency syndrome (AIDS). In this review, we describe a few examples of the mechanisms that some human viruses, that is, rabies virus, adenovirus, herpes simplex virus, human immunodeficiency virus, influenza A virus and papillomavirus, have developed to hijack dyneins, kinesins and myosins.
Collapse
Affiliation(s)
- Clàudia Río-Bergé
- Department of Biomedical Sciences of Cells & Systems, Molecular Cell Biology Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yingying Cong
- Department of Biomedical Sciences of Cells & Systems, Molecular Cell Biology Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells & Systems, Molecular Cell Biology Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
9
|
Lan J, Li W, Yu R, Syed F, Yu Q. Cell-to-cell transmission of HIV-1 from provirus-activated cells to resting naïve and memory human primary CD4 T cells is highly efficient and requires CD4 and F-actin but not chemokine receptors. J Med Virol 2022; 94:5434-5450. [PMID: 35840493 PMCID: PMC9543916 DOI: 10.1002/jmv.28005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 12/15/2022]
Abstract
Latently infected cells harboring replication-competent proviruses represent a major barrier to HIV-1 cure. One major effort to purge these cells has focused on developing the "shock and kill" approach for forcing provirus reactivation to induce cell killing by viral cytopathic effects, host immune responses, or both. We conducted kinetic and mechanistic studies of HIV-1 protein expression, virion production, and cell-to-cell virus transmission during provirus reactivation. Provirus-activated ACH-2 cells stimulated with romidepsin (RMD) or PMA produced Nef early, and then Env and Gag in parallel with the appearance of virions. Env on the surface of provirus-activated cells and cellular F-actin were critical in the formation of virological synapses to mediate cell-to-cell transmission of HIV-1 from provirus-activated cells to uninfected cells. This HIV-1 cell-to-cell transmission was substantially more efficient than transmission seen via cell-free virus spread and required F-actin remodeling and CD4, but not chemokine receptors. Resting human primary CD4+ T cells including naïve and memory subpopulations and, especially the memory CD4+ T cells, were highly susceptible to HIV-1 infection via cell-to-cell transmission. Cell-to-cell transmission of HIV-1 from provirus-activated cells was profoundly decreased by protease inhibitors (PIs) and neutralizing antibodies (nAbs) that recognize the CD4-binding site (CD4bs) such as VRC01, but not by reverse transcriptase (RT) inhibitor Emtricitabine (FTC). Therefore, our results suggest that PIs with potent blocking abilities should be used in clinical application of the "shock and kill" approach, most likely in combination with CD4bs nAbs, to prevent new HIV-1 infections.
Collapse
Affiliation(s)
- Jie Lan
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA,Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Wei Li
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Richard Yu
- Department of Internal Medicine, School of MedicineUniversity of NevadaRenoNevadaUSA
| | - Fahim Syed
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Qigui Yu
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
10
|
Screening and analysis of immune-related genes of Aedes aegypti infected with DENV2. Acta Trop 2022; 236:106698. [PMID: 36162456 DOI: 10.1016/j.actatropica.2022.106698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 01/08/2023]
Abstract
Dengue virus type Ⅱ (DENV2) is a primary serotype responsible for the dengue fever epidemic, and Aedes aegypti is the main DENV2 vector. Understanding the Aedes aegypti immune mechanism against DENV2 is the basis for research on immune blockade in mosquitoes. Some preliminary studies lack validation in the literature, so this study was performed to further study and validate the potential target genes to provide a further basis for screening key target genes. We screened 51 genes possibly related to Aedes aegypti infection and immunity from the literature for further verification. First, bioinformatic methods such as GO, KEGG and PPI analysis were used, and then RT-qPCR was used to detect the changes in mRNA expression in the midguts and salivary glands of Aedes aegypti infected with DENV2.Bioinformatic analysis showed that mostly genes of the glucose metabolism pathway and myoprotein were influenced. In salivary glands, the Gst (xa) and Toll (xb) expression levels were significantly correlated with DENV2 load (y, lg[DENV2 RNA copies]), y = -3436xa+0.2287xb+3.8194 (adjusted R2 = 0.5563, F = 9.148, PF = 0.0045). In midguts, DENV2 load was significantly correlated with the relative Fba(R2 = 0.4381, t = 2.497, p < 0.05, df = 8), UcCr(R2 = 0.4072, t = 2.344, p < 0.05, df = 8) and Gbps1(R2 = 0.4678, t = 2.652, p < 0.05, df = 8) expression levels, but multiple regression did not yield significant results. This study shows that genes related to glucose metabolism and muscle proteins contribute to the interaction between Aedes aegypti and dengue virus. It was confirmed that SAAG-4, histone H4, endoplasmin, catalase and other genes are involved in the regulation of DENV2 infection in Aedes aegypti. It was revealed that GST and Toll in salivary glands may have antagonistic effects on the regulation of DENV2 load. Fba, UcCr and Gbps1 in the midgut may increase DENV2 load. These study results further condensed the potential target gene range of the Aedes aegypti immune mechanism against DENV2 infection and provided basic information for research on the Aedes aegypti in vivo blockade strategy against DENV2.
Collapse
|
11
|
Hoffman HK, Aguilar RS, Clark AR, Groves NS, Pezeshkian N, Bruns MM, van Engelenburg SB. Endocytosed HIV-1 Envelope Glycoprotein Traffics to Rab14 + Late Endosomes and Lysosomes to Regulate Surface Levels in T-Cell Lines. J Virol 2022; 96:e0076722. [PMID: 35770989 PMCID: PMC9327703 DOI: 10.1128/jvi.00767-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/06/2022] [Indexed: 11/20/2022] Open
Abstract
Production of infectious HIV-1 particles requires incorporation of the viral envelope glycoprotein (Env) at the plasma membrane (PM) of infected CD4+ T cells. Env trafficking to the PM exposes viral epitopes that can be exploited by the host immune system; however, HIV-1 can evade this response by endocytosis of excess Env from the PM. The fate of Env after internalization remains unclear, with evidence suggesting several different vesicular trafficking steps may be involved, including recycling pathways. To date, there have been very few studies documenting the trafficking pathways of native Env in infected T cells. Furthermore, it remains unclear whether there are T-cell-specific endosomal pathways regulating the fate of endocytic Env. Here, we use a pulse-labeling approach with a monovalent anti-Env Fab probe to characterize the trafficking of internalized Env within infected CD4+ T-cell lines, together with CRISPR/Cas9-mediated endogenous protein tagging, to assess the role of host cell Rab GTPases in Env trafficking. We show that endocytosed Env traffics to Rab14+ compartments that possess hallmarks of late endosomes and lysosomes. We also demonstrate that Env can recycle back to the PM, although we find that recycling does not occur at high rates when compared to the model recycling protein transferrin. These results help to resolve open questions about the fate and relevance of endocytosed Env in HIV-infected cells and suggest a novel role for Rab14 in a cell-type-specific late-endosomal/lysosomal trafficking pathway in T cells. IMPORTANCE HIV-1 envelope glycoprotein (Env) evades immune neutralization through many mechanisms. One immune evasion strategy may result from the internalization of excess surface-exposed Env to prevent antibody-dependent cellular cytotoxicity or neutralization. Characterization of the fate of endocytosed Env is critical to understand which vesicular pathways could be targeted to promote display of Env epitopes to the immune system. In this study, we characterize the endocytic fate of native Env, expressed from infected human T-cell lines. We demonstrate that Env is rapidly trafficked to a late-endosome/lysosome-like compartment and can be recycled to the cell surface for incorporation into virus assembly sites. This study implicates a novel intracellular compartment, marked by host-cell Rab14 GTPases, for the sequestration of Env. Therapeutic approaches aimed at mobilizing this intracellular pool of Env could lead to stronger immune control of HIV-1 infection via antibody-dependent cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Huxley K. Hoffman
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Rebekah S. Aguilar
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Austin R. Clark
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Nicholas S. Groves
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Nairi Pezeshkian
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Merissa M. Bruns
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| | - Schuyler B. van Engelenburg
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, Colorado, USA
| |
Collapse
|
12
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
13
|
Abstract
The HIV Env glycoprotein is the surface glycoprotein responsible for viral entry into CD4+ immune cells. During infection, Env also serves as a primary target for antibody responses, which are robust but unable to control virus replication. Immune evasion by HIV-1 Env appears to employ complex mechanisms to regulate what antigenic states are presented to the immune system. Immunodominant features appear to be distinct from epitopes that interfere with Env functions in mediating infection. Further, cell-cell transmission studies indicate that vulnerable conformational states are additionally hidden from recognition on infected cells, even though the presence of Env at the cell surface is required for viral infection through the virological synapse. Cell-cell infection studies support that Env on infected cells is presented in distinct conformations from that on virus particles. Here we review data regarding the regulation of conformational states of Env and assess how regulated sorting of Env within the infected cell may underlie mechanisms to distinguish Env on the surface of virus particles versus Env on the surface of infected cells. These mechanisms may allow infected cells to avoid opsonization, providing cell-to-cell infection by HIV with a selective advantage during evolution within an infected individual. Understanding how distinct Env conformations are presented on cells versus viruses may be essential to designing effective vaccine approaches and therapeutic strategies to clear infected cell reservoirs.
Collapse
Affiliation(s)
- Connie Zhao
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hongru Li
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Talia H. Swartz
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benjamin K. Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
14
|
HIV-1 and HTLV-1 Transmission Modes: Mechanisms and Importance for Virus Spread. Viruses 2022; 14:v14010152. [PMID: 35062355 PMCID: PMC8779814 DOI: 10.3390/v14010152] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
So far, only two retroviruses, human immunodeficiency virus (HIV) (type 1 and 2) and human T-cell lymphotropic virus type 1 (HTLV-1), have been recognized as pathogenic for humans. Both viruses mainly infect CD4+ T lymphocytes. HIV replication induces the apoptosis of CD4 lymphocytes, leading to the development of acquired immunodeficiency syndrome (AIDS). After a long clinical latency period, HTLV-1 can transform lymphocytes, with subsequent uncontrolled proliferation and the manifestation of a disease called adult T-cell leukemia (ATLL). Certain infected patients develop neurological autoimmune disorder called HTLV-1-associated myelopathy, also known as tropical spastic paraparesis (HAM/TSP). Both viruses are transmitted between individuals via blood transfusion, tissue/organ transplantation, breastfeeding, and sexual intercourse. Within the host, these viruses can spread utilizing either cell-free or cell-to-cell modes of transmission. In this review, we discuss the mechanisms and importance of each mode of transmission for the biology of HIV-1 and HTLV-1.
Collapse
|
15
|
Snetkov X, Haider T, Mesner D, Groves N, van Engelenburg SB, Jolly C. A Conserved Tryptophan in the Envelope Cytoplasmic Tail Regulates HIV-1 Assembly and Spread. Viruses 2022; 14:v14010129. [PMID: 35062333 PMCID: PMC8778169 DOI: 10.3390/v14010129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
The HIV-1 envelope (Env) is an essential determinant of viral infectivity, tropism and spread between T cells. Lentiviral Env contain an unusually long 150 amino acid cytoplasmic tail (EnvCT), but the function of the EnvCT and many conserved domains within it remain largely uncharacterised. Here, we identified a highly conserved tryptophan motif at position 757 (W757) in the LLP-2 alpha helix of the EnvCT as a key determinant for HIV-1 replication and spread between T cells. Alanine substitution at this position potently inhibited HIV-1 cell–cell spread (the dominant mode of HIV-1 dissemination) by preventing recruitment of Env and Gag to sites of cell–cell contact, inhibiting virological synapse (VS) formation and spreading infection. Single-molecule tracking and super-resolution imaging showed that mutation of W757 dysregulates Env diffusion in the plasma membrane and increases Env mobility. Further analysis of Env function revealed that W757 is also required for Env fusion and infectivity, which together with reduced VS formation, result in a potent defect in viral spread. Notably, W757 lies within a region of the EnvCT recently shown to act as a supporting baseplate for Env. Our data support a model in which W757 plays a key role in regulating Env biology, modulating its temporal and spatial recruitment to virus assembly sites and regulating the inherent fusogenicity of the Env ectodomain, thereby supporting efficient HIV-1 replication and spread.
Collapse
Affiliation(s)
- Xenia Snetkov
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; (X.S.); (T.H.); (D.M.)
| | - Tafhima Haider
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; (X.S.); (T.H.); (D.M.)
| | - Dejan Mesner
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; (X.S.); (T.H.); (D.M.)
| | - Nicholas Groves
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, CO 80210, USA; (N.G.); (S.B.v.E.)
| | - Schuyler B. van Engelenburg
- Molecular and Cellular Biophysics Program, Department of Biological Sciences, University of Denver, Denver, CO 80210, USA; (N.G.); (S.B.v.E.)
| | - Clare Jolly
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; (X.S.); (T.H.); (D.M.)
- Correspondence:
| |
Collapse
|
16
|
Perez-Zsolt D, Raïch-Regué D, Muñoz-Basagoiti J, Aguilar-Gurrieri C, Clotet B, Blanco J, Izquierdo-Useros N. HIV-1 trans-Infection Mediated by DCs: The Tip of the Iceberg of Cell-to-Cell Viral Transmission. Pathogens 2021; 11:39. [PMID: 35055987 PMCID: PMC8778849 DOI: 10.3390/pathogens11010039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
HIV-1 cell-to-cell transmission is key for an effective viral replication that evades immunity. This highly infectious mechanism is orchestrated by different cellular targets that utilize a wide variety of processes to efficiently transfer HIV-1 particles. Dendritic cells (DCs) are the most potent antigen presenting cells that initiate antiviral immune responses, but are also the cells with highest capacity to transfer HIV-1. This mechanism, known as trans-infection, relies on the capacity of DCs to capture HIV-1 particles via lectin receptors such as the sialic acid-binding I-type lectin Siglec-1/CD169. The discovery of the molecular interaction of Siglec-1 with sialylated lipids exposed on HIV-1 membranes has enlightened how this receptor can bind to several enveloped viruses. The outcome of these interactions can either mount effective immune responses, boost the productive infection of DCs and favour innate sensing, or fuel viral transmission via trans-infection. Here we review these scenarios focusing on HIV-1 and other enveloped viruses such as Ebola virus or SARS-CoV-2.
Collapse
Affiliation(s)
- Daniel Perez-Zsolt
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916 Badalona, Spain; (D.P.-Z.); (D.R.-R.); (J.M.-B.); (C.A.-G.); (B.C.); (J.B.)
| | - Dàlia Raïch-Regué
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916 Badalona, Spain; (D.P.-Z.); (D.R.-R.); (J.M.-B.); (C.A.-G.); (B.C.); (J.B.)
| | - Jordana Muñoz-Basagoiti
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916 Badalona, Spain; (D.P.-Z.); (D.R.-R.); (J.M.-B.); (C.A.-G.); (B.C.); (J.B.)
| | - Carmen Aguilar-Gurrieri
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916 Badalona, Spain; (D.P.-Z.); (D.R.-R.); (J.M.-B.); (C.A.-G.); (B.C.); (J.B.)
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916 Badalona, Spain; (D.P.-Z.); (D.R.-R.); (J.M.-B.); (C.A.-G.); (B.C.); (J.B.)
- Infectious Diseases and Immunity Department, Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916 Badalona, Spain; (D.P.-Z.); (D.R.-R.); (J.M.-B.); (C.A.-G.); (B.C.); (J.B.)
- Infectious Diseases and Immunity Department, Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Can Ruti Campus, 08916 Badalona, Spain; (D.P.-Z.); (D.R.-R.); (J.M.-B.); (C.A.-G.); (B.C.); (J.B.)
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain
| |
Collapse
|
17
|
Post-translational modifications and stabilization of microtubules regulate transport of viral factors during infections. Biochem Soc Trans 2021; 49:1735-1748. [PMID: 34436545 DOI: 10.1042/bst20210017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/17/2022]
Abstract
Tubulin post-translational modifications (PTMs) constitute a source of diversity for microtubule (MT) functions, in addition to the different isotypes of α and β-tubulin acting as building blocks of MTs. Also, MT-associated proteins (MAPs) confer different characteristics to MTs. The combination of all these factors regulates the stability of these structures that act as rails to transport organelles within the cell, facilitating the association of motor complexes. All these functions are involved in crucial cellular processes in most cell types, ranging from spindle formation in mitosis to the defense against incoming cellular threats during phagocytosis mediated by immune cells. The regulation of MT dynamics through tubulin PTMs has evolved to depend on many different factors that act in a complex orchestrated manner. These tightly regulated processes are particularly relevant during the induction of effective immune responses against pathogens. Viruses have proved not only to hijack MTs and MAPs in order to favor an efficient infection, but also to induce certain PTMs that improve their cellular spread and lead to secondary consequences of viral processes. In this review, we offer a perspective on relevant MT-related elements exploited by viruses.
Collapse
|
18
|
Abstract
The cellular surfaceome and its residing extracellularly exposed proteins are involved in a multitude of molecular signaling processes across the viral infection cycle. Successful viral propagation, including viral entry, immune evasion, virion release and viral spread rely on dynamic molecular interactions with the surfaceome. Decoding of these viral-host surfaceome interactions using advanced technologies enabled the discovery of fundamental new functional insights into cellular and viral biology. In this review, we highlight recently developed experimental strategies, with a focus on spatial proteotyping technologies, aiding in the rational design of theranostic strategies to combat viral infections.
Collapse
|
19
|
Zandi F, Khalaj V, Goshadrou F, Meyfour A, Gholami A, Enayati S, Mehranfar M, Rahmati S, Kheiri EV, Badie HG, Vaziri B. Rabies virus matrix protein targets host actin cytoskeleton: a protein-protein interaction analysis. Pathog Dis 2020; 79:6027507. [PMID: 33289839 DOI: 10.1093/femspd/ftaa075] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Multifunctional matrix protein (M) of rabies virus (RABV) plays essential roles in the pathogenesis of rabies infection. Identification of M protein interacting partners in target hosts could help to elucidate the biological pathways and molecular mechanisms involved in the pathogenesis of this virus. In this study, two-dimensional Far-western blotting (2D-Far-WB) technique was applied to find possible matrix protein partners in the rat brainstem. Recombinant RABV M was expressed in Pichia pastoris and was partially purified. Subsequently, 2D-Far-WB-determined six rat brainstem proteins interacted with recombinant M proteins that were identified by mass spectrometry. Functional annotation by gene ontology analysis determined these proteins were involved in the regulation of synaptic transmission processes, metabolic process and cell morphogenesis-cytoskeleton organization. The interaction of viral M protein with selected host proteins in mouse Neuro-2a cells infected with RABV was verified by super-resolution confocal microscopy. Molecular docking simulations also demonstrated the formation of RABV M complexes. However, further confirmation with co-immunoprecipitation was only successful for M-actin cytoplasmic 1 interaction. Our study revealed actin cytoplasmic 1 as a binding partner of M protein, which might have important role(s) in rabies pathogenesis.
Collapse
Affiliation(s)
- Fatemeh Zandi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.,Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, 1971653313, Iran
| | - Vahid Khalaj
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Goshadrou
- Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, 1971653313, Iran
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717413, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, 16635-148, Iran
| | - Alireza Gholami
- Department of Virology, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Somayeh Enayati
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahsa Mehranfar
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Saman Rahmati
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | | | - Hamid Gholamipour Badie
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Behrouz Vaziri
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| |
Collapse
|
20
|
Lyu Y, Kopcho S, Mohan M, Okeoma CM. Long-Term Low-Dose Delta-9-Tetrahydrocannbinol (THC) Administration to Simian Immunodeficiency Virus (SIV) Infected Rhesus Macaques Stimulates the Release of Bioactive Blood Extracellular Vesicles (EVs) that Induce Divergent Structural Adaptations and Signaling Cues. Cells 2020; 9:E2243. [PMID: 33036231 PMCID: PMC7599525 DOI: 10.3390/cells9102243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022] Open
Abstract
Blood extracellular vesicles (BEVs) carry bioactive cargo (proteins, genetic materials, lipids, licit, and illicit drugs) that regulate diverse functions in target cells. The cannabinoid drug delta-9-tetrahydrocannabinol (THC) is FDA approved for the treatment of anorexia and weight loss in people living with HIV. However, the effect of THC on BEV characteristics in the setting of HIV/SIV infection needs to be determined. Here, we used the SIV-infected rhesus macaque model of AIDS to evaluate the longitudinal effects of THC (THC/SIV) or vehicle (VEH/SIV) treatment in HIV/SIV infection on the properties of BEVs. While BEV concentrations increased longitudinally (pre-SIV (0), 30, and 150 days post-SIV infection (DPI)) in VEH/SIV macaques, the opposite trend was observed with THC/SIV macaques. SIV infection altered BEV membrane properties and cargo composition late in infection, since i) the electrostatic surface properties (zeta potential, ζ potential) showed that RM BEVs carried negative surface charge, but at 150 DPI, SIV infection significantly changed BEV ζ potential; ii) BEVs from the VEH/SIV group altered tetraspanin CD9 and CD81 levels compared to the THC/SIV group. Furthermore, VEH/SIV and THC/SIV BEVs mediated divergent changes in monocyte gene expression, morphometrics, signaling, and function. These include altered tetraspanin and integrin β1 expression; altered levels and distribution of polymerized actin, FAK/pY397 FAK, pERK1/2, cleaved caspase 3, proapoptotic Bid and truncated tBid; and altered adhesion of monocytes to collagen I. These data indicate that HIV/SIV infection and THC treatment result in the release of bioactive BEVs with potential to induce distinct structural adaptations and signaling cues to instruct divergent cellular responses to infection.
Collapse
Affiliation(s)
- Yuan Lyu
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794-8651, USA; (Y.L.); (S.K.)
| | - Steven Kopcho
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794-8651, USA; (Y.L.); (S.K.)
| | - Mahesh Mohan
- Host Pathogen Interaction, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227-5302, USA
| | - Chioma M. Okeoma
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794-8651, USA; (Y.L.); (S.K.)
| |
Collapse
|
21
|
Schwartz D, Iyengar S. Recognition of Apoptotic Cells by Viruses and Cytolytic Lymphocytes: Target Selection in the Fog of War. Viral Immunol 2020; 33:188-196. [PMID: 32286181 PMCID: PMC7185367 DOI: 10.1089/vim.2019.0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Viruses and cytolytic lymphocytes operate in an environment filled with dying and dead cells, and cell fragments. For viruses, irreversible fusion with doomed cells is suicide. For cytotoxic T lymphocyte and natural killer cells, time and limited lytic resources spent on apoptotic targets is wasteful and may result in death of the host. We make the case that the target membrane cytoskeleton is the best source of information regarding the suitability of potential targets for engagement for both viruses and lytic effector cells, and we present experimental evidence for detection of apoptotic cells by HIV, without loss of infectivity.
Collapse
Affiliation(s)
- David Schwartz
- Jurist Research Department, Hackensack University Medical Center, Hackensack, New Jersey
| | - Sujatha Iyengar
- Jurist Research Department, Hackensack University Medical Center, Hackensack, New Jersey
| |
Collapse
|
22
|
Loss of Nef-mediated CD3 down-regulation in the HIV-1 lineage increases viral infectivity and spread. Proc Natl Acad Sci U S A 2020; 117:7382-7391. [PMID: 32179688 PMCID: PMC7132320 DOI: 10.1073/pnas.1921135117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Lentiviruses encode accessory proteins to manipulate their host cells in order to efficiently replicate and evade antiviral defenses. Interestingly, most lentiviral Nefs down-regulate CD3 from the surface of infected T cells to perturb immune responses. However, for reasons that are incompletely understood, HIV-1 and its simian immunodeficiency virus ancestors lack this function. Here, we report that engineering HIV-1 for Nef-mediated down-regulation of CD3 reduces Env-dependent HIV-1 infectivity, resulting in less efficient cell-to-cell spread and replication. Our data suggest that HIV-1 may have evolved to lose the CD3 down-modulation function of Nef in order to allow T cell activation and to boost viral replication, possibly at the cost of less effective immune evasion and increased pathogenicity. Nef is an accessory protein of primate lentiviruses that is essential for efficient replication and pathogenesis of HIV-1. A conserved feature of Nef proteins from different lentiviral lineages is the ability to modulate host protein trafficking and down-regulate a number of cell surface receptors to enhance replication and promote immune evasion. Notably, the inability of Nef to down-regulate CD3 from infected T cells distinguishes HIV-1 Nef and its direct simian precursors from other primate lentiviruses. Why HIV-1 does not employ this potential immune evasion strategy is not fully understood. Using chimeric HIV-1 constructs expressing lentiviral Nef proteins that differ in their ability to down-modulate CD3, we show that retaining CD3 on the surface of infected primary T cells results in increased viral replication and cell-to-cell spread. We identified increased expression of envelope (Env) trimers at the cell surface and increased Env incorporation into virions as the determinants for the Nef- and CD3-dependent enhancement of viral infectivity. Importantly, this was independent of Nef-mediated antagonism of the host restriction factor SERINC5. CD3 retention on the surface of infected primary T cells also correlated with increased T cell signaling, activation, and cell death during cell-to-cell spread. Taken together, our results show that loss of an otherwise conserved function of Nef has a positive effect on HIV-1 replication, allowing for more efficient replication while potentially contributing to HIV-1 pathogenesis by triggering T cell activation and cell death during viral spread.
Collapse
|
23
|
Rodríguez-Gallego E, Tarancón-Diez L, García F, Del Romero J, Benito JM, Alba V, Herrero P, Rull A, Dominguez-Molina B, Martinez-Madrid O, Martin-Pena L, Pulido F, León A, Rodríguez C, Rallón N, Peraire J, Viladés C, Leal M, Vidal F, Ruiz-Mateos E. Proteomic Profile Associated With Loss of Spontaneous Human Immunodeficiency Virus Type 1 Elite Control. J Infect Dis 2020; 219:867-876. [PMID: 30312441 DOI: 10.1093/infdis/jiy599] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Elite controllers (ECs) spontaneously control plasma human immunodeficiency virus type 1 (HIV-1) RNA without antiretroviral therapy. However, 25% lose virological control over time. The aim of this work was to study the proteomic profile that preceded this loss of virological control to identify potential biomarkers. METHODS Plasma samples from ECs who spontaneously lost virological control (transient controllers [TCs]), at 2 years and 1 year before the loss of control, were compared with a control group of ECs who persistently maintained virological control during the same follow-up period (persistent controllers [PCs]). Comparative plasma shotgun proteomics was performed with tandem mass tag (TMT) isobaric tag labeling and nanoflow liquid chromatography coupled to Orbitrap mass spectrometry. RESULTS Eighteen proteins exhibited differences comparing PC and preloss TC timepoints. These proteins were involved in proinflammatory mechanisms, and some of them play a role in HIV-1 replication and pathogenesis and interact with structural viral proteins. Coagulation factor XI, α-1-antichymotrypsin, ficolin-2, 14-3-3 protein, and galectin-3-binding protein were considered potential biomarkers. CONCLUSIONS The proteomic signature associated with the spontaneous loss of virological control was characterized by higher levels of inflammation, transendothelial migration, and coagulation. Galectin-3 binding protein could be considered as potential biomarker for the prediction of virological progression and as therapeutic target in ECs.
Collapse
Affiliation(s)
- Esther Rodríguez-Gallego
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona
| | - Laura Tarancón-Diez
- Laboratory of Immunovirology, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/Consejo Superior de Investigaciones Científicas/University of Seville, Spain
| | - Felipe García
- Hospital Clinic-Fundació Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centre Català d'Investigació i Desenvolupament de Vacunes contra la Sida, Universidad de Barcelona, Spain
| | - Jorge Del Romero
- Centro Sanitario Sandoval, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Jose Miguel Benito
- IIS-Fundación Jiménez Diaz, Universidad Autónoma de Madrid/Madrid Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - Verónica Alba
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona
| | - Pol Herrero
- Centre for Omic Sciences, Unitat Mixta Universitat Rovira i Virgili-Eurecat, Reus, Spain
| | - Anna Rull
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona
| | - Beatriz Dominguez-Molina
- Laboratory of Immunovirology, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/Consejo Superior de Investigaciones Científicas/University of Seville, Spain
| | - Onofre Martinez-Madrid
- Unidad Enfermedades Infecciosas, Hospital General Universitario Santa Lucía, Cartagena, Spain
| | - Luisa Martin-Pena
- Infectious Disease Service, Son Espases Hospital, Palma de Mallorca, Illes Balears, Spain.,Multidisciplinary Group for Infectious Disease Service, Institute of Health Sciences Research, Instituto de Investigación Sanitaria de Palma, Health Research Foundation Ramón Llull, Son Espases Hospital, Palma de Mallorca, Illes Balears
| | - Federico Pulido
- HIV Unit, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - Agathe León
- Hospital Clinic-Fundació Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centre Català d'Investigació i Desenvolupament de Vacunes contra la Sida, Universidad de Barcelona, Spain
| | - Carmen Rodríguez
- Centro Sanitario Sandoval, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Norma Rallón
- IIS-Fundación Jiménez Diaz, Universidad Autónoma de Madrid/Madrid Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - Joaquim Peraire
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona
| | - Consuelo Viladés
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona
| | - Manuel Leal
- Laboratory of Immunovirology, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/Consejo Superior de Investigaciones Científicas/University of Seville, Spain.,Servicio de Medicina Interna, Hospital Viamed Santa Ángela de la Cruz, Sevilla, Spain
| | - Francesc Vidal
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona
| | - Ezequiel Ruiz-Mateos
- Laboratory of Immunovirology, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/Consejo Superior de Investigaciones Científicas/University of Seville, Spain
| | | |
Collapse
|
24
|
Lundberg M, Mattsson Å, Reiser K, Holmgren A, Curbo S. Inhibition of the thioredoxin system by PX-12 (1-methylpropyl 2-imidazolyl disulfide) impedes HIV-1 infection in TZM-bl cells. Sci Rep 2019; 9:5656. [PMID: 30948772 PMCID: PMC6449384 DOI: 10.1038/s41598-019-42068-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/25/2019] [Indexed: 12/18/2022] Open
Abstract
Human immunodeficiency virus (HIV-1) entry is initiated by the binding between the viral envelope glycoprotein gp120 and the host receptor CD4, and followed by reduction of structural disulfides of gp120 and CD4. The host thioredoxin-1 (Trx1) efficiently reduces disulfides of gp120 and CD4 in vitro, and recently CD4-dependent HIV-1 entry was shown to be inhibited by anti-Trx1-antibodies, indicating a central role for Trx1. 1-methylpropyl-2-imidazolyl disulfide (PX-12) is a reversible inhibitor of the Trx1 system that may also cause a slow irreversible thioalkylation of Trx1. It was developed as an antitumor agent, however, the current study aimed to determine if it also has an anti-HIV-1 effect. We show that PX-12 has anti-HIV-1(IIIB) activity in TZM-bl cells, in fact, no virus was detected inside the cells in the presence of 10 µM PX-12. Moreover, PX-12 inhibited the enzymatic activity of Trx1 and the Trx1-dependent disulfide reduction of gp120. Microtubule polymerization and formation of acetylated microtubules were also inhibited, activities shown to be required for HIV-1 life cycle propagation. In conclusion, our data strengthens the notion that the early steps of the HIV-1 life cycle depends on the Trx1 system and indicate that the Trx1 system may be a rational drug target for HIV-1 treatment.
Collapse
Affiliation(s)
- Mathias Lundberg
- Department of Clinical Science and Education, Södersjukhuset, Internal medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Åse Mattsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Kathrin Reiser
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Arne Holmgren
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Sophie Curbo
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
The Influence of Envelope C-Terminus Amino Acid Composition on the Ratio of Cell-Free to Cell-Cell Transmission for Bovine Foamy Virus. Viruses 2019; 11:v11020130. [PMID: 30708993 PMCID: PMC6410131 DOI: 10.3390/v11020130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 12/13/2022] Open
Abstract
Foamy viruses (FVs) have extensive cell tropism in vitro, special replication features, and no clinical pathogenicity in naturally or experimentally infected animals, which distinguish them from orthoretroviruses. Among FVs, bovine foamy virus (BFV) has undetectable or extremely low levels of cell-free transmission in the supernatants of infected cells and mainly spreads by cell-to-cell transmission, which deters its use as a gene transfer vector. Here, using an in vitro virus evolution system, we successfully isolated high-titer cell-free BFV strains from the original cell-to-cell transmissible BFV3026 strain and further constructed an infectious cell-free BFV clone called pBS-BFV-Z1. Following sequence alignment with a cell-associated clone pBS-BFV-B, we identified a number of changes in the genome of pBS-BFV-Z1. Extensive mutagenesis analysis revealed that the C-terminus of envelope protein, especially the K898 residue, controls BFV cell-free transmission by enhancing cell-free virus entry but not the virus release capacity. Taken together, our data show the genetic determinants that regulate cell-to-cell and cell-free transmission of BFV.
Collapse
|
26
|
Bayliss RJ, Piguet V. Masters of manipulation: Viral modulation of the immunological synapse. Cell Microbiol 2018; 20:e12944. [PMID: 30123959 PMCID: PMC6492149 DOI: 10.1111/cmi.12944] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/01/2018] [Accepted: 08/14/2018] [Indexed: 02/06/2023]
Abstract
In order to thrive, viruses have evolved to manipulate host cell machinery for their own benefit. One major obstacle faced by pathogens is the immunological synapse. To enable efficient replication and latency in immune cells, viruses have developed a range of strategies to manipulate cellular processes involved in immunological synapse formation to evade immune detection and control T-cell activation. In vitro, viruses such as human immunodeficiency virus 1 and human T-lymphotropic virus type 1 utilise structures known as virological synapses to aid transmission of viral particles from cell to cell in a process termed trans-infection. The formation of the virological synapse provides a gateway for virus to be transferred between cells avoiding the extracellular space, preventing antibody neutralisation or recognition by complement. This review looks at how viruses are able to subvert intracellular signalling to modulate immune function to their advantage and explores the role synapse formation has in viral persistence and cell-to-cell transmission.
Collapse
Affiliation(s)
- Rebecca J. Bayliss
- Division of Infection and Immunity, School of MedicineCardiff UniversityCardiffUK
| | - Vincent Piguet
- Division of Infection and Immunity, School of MedicineCardiff UniversityCardiffUK
- Division of Dermatology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Division of DermatologyWomen's College HospitalTorontoOntarioCanada
| |
Collapse
|
27
|
Chen L, Keppler OT, Schölz C. Post-translational Modification-Based Regulation of HIV Replication. Front Microbiol 2018; 9:2131. [PMID: 30254620 PMCID: PMC6141784 DOI: 10.3389/fmicb.2018.02131] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022] Open
Abstract
Human immunodeficiency virus (HIV) relies heavily on the host cellular machinery for production of viral progeny. To exploit cellular proteins for replication and to overcome host factors with antiviral activity, HIV has evolved a set of regulatory and accessory proteins to shape an optimized environment for its replication and to facilitate evasion from the immune system. Several cellular pathways are hijacked by the virus to modulate critical steps during the viral life cycle. Thereby, post-translational modifications (PTMs) of viral and cellular proteins gain increasingly attention as modifying enzymes regulate virtually every step of the viral replication cycle. This review summarizes the current knowledge of HIV-host interactions influenced by PTMs with a special focus on acetylation, ubiquitination, and phosphorylation of proteins linked to cellular signaling and viral replication. Insights into these interactions are surmised to aid development of new intervention strategies.
Collapse
Affiliation(s)
- Lin Chen
- Max von Pettenkofer-Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver T Keppler
- Max von Pettenkofer-Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Schölz
- Max von Pettenkofer-Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
28
|
Kamiyama H, Izumida M, Umemura Y, Hayashi H, Matsuyama T, Kubo Y. Role of Ezrin Phosphorylation in HIV-1 Replication. Front Microbiol 2018; 9:1912. [PMID: 30210460 PMCID: PMC6119696 DOI: 10.3389/fmicb.2018.01912] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/30/2018] [Indexed: 11/15/2022] Open
Abstract
Host-cell expression of the ezrin protein is required for CXCR4 (X4)-tropic HIV-1 infection. Ezrin function is regulated by phosphorylation at threonine-567. This study investigates the role of ezrin phosphorylation in HIV-1 infection and virion release. We analyzed the effects of ezrin mutations involving substitution of threonine-567 by alanine (EZ-TA), a constitutively inactive mutant, or by aspartic acid (EZ-TD), which mimics phosphorylated threonine. We also investigated the effects of ezrin silencing on HIV-1 virion release using a specific siRNA. We observed that X4-tropic HIV-1 vector infection was inhibited by expression of the EZ-TA mutant but increased by expression of the EZ-TD mutant, suggesting that ezrin phosphorylation in target cells is required for efficient HIV-1 entry. Expression of a dominant-negative mutant of ezrin (EZ-N) and ezrin silencing in HIV-1 vector-producing cells significantly reduced the infectivity of released virions without affecting virion production. This result indicates that endogenous ezrin expression is required for virion infectivity. The EZ-TD but not the EZ-TA inhibited virion release from HIV-1 vector-producing cells. Taken together, these findings suggest that ezrin phosphorylation in target cells is required for efficient HIV-1 entry but inhibits virion release from HIV-1 vector-producing cells.
Collapse
Affiliation(s)
- Haruka Kamiyama
- Department of AIDS Research, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.,Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Mai Izumida
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Yuria Umemura
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Hideki Hayashi
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Medical University Research Administrator (MEDURA), Nagasaki University School of Medicine, Nagasaki, Japan
| | - Toshifumi Matsuyama
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Department of Cancer Stem Cell Biology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yoshinao Kubo
- Department of AIDS Research, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.,Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
29
|
Dufloo J, Bruel T, Schwartz O. HIV-1 cell-to-cell transmission and broadly neutralizing antibodies. Retrovirology 2018; 15:51. [PMID: 30055632 PMCID: PMC6064125 DOI: 10.1186/s12977-018-0434-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022] Open
Abstract
HIV-1 spreads through contacts between infected and target cells. Polarized viral budding at the contact site forms the virological synapse. Additional cellular processes, such as nanotubes, filopodia, virus accumulation in endocytic or phagocytic compartments promote efficient viral propagation. Cell-to-cell transmission allows immune evasion and likely contributes to HIV-1 spread in vivo. Anti-HIV-1 broadly neutralizing antibodies (bNAbs) defeat the majority of circulating viral strains by binding to the viral envelope glycoprotein (Env). Several bNAbs have entered clinical evaluation during the last years. It is thus important to understand their mechanism of action and to determine how they interact with infected cells. In experimental models, HIV-1 cell-to-cell transmission is sensitive to neutralization, but the effect of antibodies is often less marked than during cell-free infection. This may be due to differences in the conformation or accessibility of Env at the surface of virions and cells. In this review, we summarize the current knowledge on HIV-1 cell-to-cell transmission and discuss the role of bNAbs during this process.
Collapse
Affiliation(s)
- Jérémy Dufloo
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, Paris, France.,CNRS-UMR3569, Paris, France
| | - Timothée Bruel
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, Paris, France.,CNRS-UMR3569, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, Paris, France. .,CNRS-UMR3569, Paris, France. .,Vaccine Research Institute, Créteil, France.
| |
Collapse
|
30
|
|
31
|
Narasimhulu VGS, Bellamy-McIntyre AK, Laumaea AE, Lay CS, Harrison DN, King HAD, Drummer HE, Poumbourios P. Distinct functions for the membrane-proximal ectodomain region (MPER) of HIV-1 gp41 in cell-free and cell-cell viral transmission and cell-cell fusion. J Biol Chem 2018; 293:6099-6120. [PMID: 29496992 DOI: 10.1074/jbc.ra117.000537] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/21/2018] [Indexed: 11/06/2022] Open
Abstract
HIV-1 is spread by cell-free virions and by cell-cell viral transfer. We asked whether the structure and function of a broad neutralizing antibody (bNAb) epitope, the membrane-proximal ectodomain region (MPER) of the viral gp41 transmembrane glycoprotein, differ in cell-free and cell-cell-transmitted viruses and whether this difference could be related to Ab neutralization sensitivity. Whereas cell-free viruses bearing W666A and I675A substitutions in the MPER lacked infectivity, cell-associated mutant viruses were able to initiate robust spreading infection. Infectivity was restored to cell-free viruses by additional substitutions in the cytoplasmic tail (CT) of gp41 known to disrupt interactions with the viral matrix protein. We observed contrasting effects on cell-free virus infectivity when W666A was introduced to two transmitted/founder isolates, but both mutants could still mediate cell-cell spread. Domain swapping indicated that the disparate W666A phenotypes of the cell-free transmitted/founder viruses are controlled by sequences in variable regions 1, 2, and 4 of gp120. The sequential passaging of an MPER mutant (W672A) in peripheral blood mononuclear cells enabled selection of viral revertants with loss-of-glycan suppressor mutations in variable region 1, suggesting a functional interaction between variable region 1 and the MPER. An MPER-directed bNAb neutralized cell-free virus but not cell-cell viral spread. Our results suggest that the MPER of cell-cell-transmitted virions has a malleable structure that tolerates mutagenic disruption but is not accessible to bNAbs. In cell-free virions, interactions mediated by the CT impose an alternative MPER structure that is less tolerant of mutagenic alteration and is efficiently targeted by bNAbs.
Collapse
Affiliation(s)
- Vani G S Narasimhulu
- From the Virus Entry and Vaccines Laboratory, Burnet Institute, Melbourne, Victoria 3004.,the Department of Microbiology and Immunology at the Peter Doherty Institute, University of Melbourne, Parkville, Victoria 3010, and
| | - Anna K Bellamy-McIntyre
- From the Virus Entry and Vaccines Laboratory, Burnet Institute, Melbourne, Victoria 3004.,the Departments of Microbiology and
| | - Annamarie E Laumaea
- From the Virus Entry and Vaccines Laboratory, Burnet Institute, Melbourne, Victoria 3004.,the Department of Microbiology and Immunology at the Peter Doherty Institute, University of Melbourne, Parkville, Victoria 3010, and
| | - Chan-Sien Lay
- Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - David N Harrison
- From the Virus Entry and Vaccines Laboratory, Burnet Institute, Melbourne, Victoria 3004
| | - Hannah A D King
- From the Virus Entry and Vaccines Laboratory, Burnet Institute, Melbourne, Victoria 3004.,the Department of Microbiology and Immunology at the Peter Doherty Institute, University of Melbourne, Parkville, Victoria 3010, and
| | - Heidi E Drummer
- From the Virus Entry and Vaccines Laboratory, Burnet Institute, Melbourne, Victoria 3004.,the Department of Microbiology and Immunology at the Peter Doherty Institute, University of Melbourne, Parkville, Victoria 3010, and.,the Departments of Microbiology and
| | - Pantelis Poumbourios
- From the Virus Entry and Vaccines Laboratory, Burnet Institute, Melbourne, Victoria 3004, .,the Departments of Microbiology and.,Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
32
|
Abstract
While HIV-1 infection of target cells with cell-free viral particles has been largely documented, intercellular transmission through direct cell-to-cell contact may be a predominant mode of propagation in host. To spread, HIV-1 infects cells of the immune system and takes advantage of their specific particularities and functions. Subversion of intercellular communication allows to improve HIV-1 replication through a multiplicity of intercellular structures and membrane protrusions, like tunneling nanotubes, filopodia, or lamellipodia-like structures involved in the formation of the virological synapse. Other features of immune cells, like the immunological synapse or the phagocytosis of infected cells are hijacked by HIV-1 and used as gateways to infect target cells. Finally, HIV-1 reuses its fusogenic capacity to provoke fusion between infected donor cells and target cells, and to form infected syncytia with high capacity of viral production and improved capacities of motility or survival. All these modes of cell-to-cell transfer are now considered as viral mechanisms to escape immune system and antiretroviral therapies, and could be involved in the establishment of persistent virus reservoirs in different host tissues.
Collapse
Affiliation(s)
- Lucie Bracq
- Inserm U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France.,International Associated Laboratory (LIA VirHost), Institut Pasteur Shanghai-Chinese Academy of Sciences, Shanghai, China.,International Associated Laboratory (LIA VirHost), CNRS, Université Paris-Descartes, Institut Pasteur, Paris, France
| | - Maorong Xie
- Inserm U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France.,International Associated Laboratory (LIA VirHost), CNRS, Université Paris-Descartes, Institut Pasteur, Paris, France
| | - Serge Benichou
- Inserm U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France.,International Associated Laboratory (LIA VirHost), Institut Pasteur Shanghai-Chinese Academy of Sciences, Shanghai, China.,International Associated Laboratory (LIA VirHost), CNRS, Université Paris-Descartes, Institut Pasteur, Paris, France
| | - Jérôme Bouchet
- Inserm U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris-Descartes, Sorbonne Paris-Cité, Paris, France.,International Associated Laboratory (LIA VirHost), CNRS, Université Paris-Descartes, Institut Pasteur, Paris, France
| |
Collapse
|
33
|
Ospina Stella A, Turville S. All-Round Manipulation of the Actin Cytoskeleton by HIV. Viruses 2018; 10:v10020063. [PMID: 29401736 PMCID: PMC5850370 DOI: 10.3390/v10020063] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 12/21/2022] Open
Abstract
While significant progress has been made in terms of human immunodeficiency virus (HIV) therapy, treatment does not represent a cure and remains inaccessible to many people living with HIV. Continued mechanistic research into the viral life cycle and its intersection with many aspects of cellular biology are not only fundamental in the continued fight against HIV, but also provide many key observations of the workings of our immune system. Decades of HIV research have testified to the integral role of the actin cytoskeleton in both establishing and spreading the infection. Here, we review how the virus uses different strategies to manipulate cellular actin networks and increase the efficiency of various stages of its life cycle. While some HIV proteins seem able to bind to actin filaments directly, subversion of the cytoskeleton occurs indirectly by exploiting the power of actin regulatory proteins, which are corrupted at multiple levels. Furthermore, this manipulation is not restricted to a discrete class of proteins, but rather extends throughout all layers of the cytoskeleton. We discuss prominent examples of actin regulators that are exploited, neutralized or hijacked by the virus, and address how their coordinated deregulation can lead to changes in cellular behavior that promote viral spreading.
Collapse
Affiliation(s)
- Alberto Ospina Stella
- The Kirby Institute, University of New South Wales (UNSW), Sydney NSW 2052, Australia.
| | - Stuart Turville
- The Kirby Institute, University of New South Wales (UNSW), Sydney NSW 2052, Australia.
| |
Collapse
|
34
|
Too IHK, Bonne I, Tan EL, Chu JJH, Alonso S. Prohibitin plays a critical role in Enterovirus 71 neuropathogenesis. PLoS Pathog 2018; 14:e1006778. [PMID: 29324904 PMCID: PMC5764453 DOI: 10.1371/journal.ppat.1006778] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022] Open
Abstract
A close relative of poliovirus, enterovirus 71 (EV71) is regarded as an important neurotropic virus of serious public health concern. EV71 causes Hand, Foot and Mouth Disease and has been associated with neurological complications in young children. Our limited understanding of the mechanisms involved in its neuropathogenesis has hampered the development of effective therapeutic options. Here, using a two-dimensional proteomics approach combined with mass spectrometry, we have identified a unique panel of host proteins that were differentially and dynamically modulated during EV71 infection of motor-neuron NSC-34 cells, which are found at the neuromuscular junctions where EV71 is believed to enter the central nervous system. Meta-analysis with previously published proteomics studies in neuroblastoma or muscle cell lines revealed minimal overlapping which suggests unique host-pathogen interactions in NSC-34 cells. Among the candidate proteins, we focused our attention on prohibitin (PHB), a protein that is involved in multiple cellular functions and the target of anti-cancer drug Rocaglamide (Roc-A). We demonstrated that cell surface-expressed PHB is involved in EV71 entry into neuronal cells specifically, while membrane-bound mitochondrial PHB associates with the virus replication complex and facilitates viral replication. Furthermore, Roc-A treatment of EV71-infected neuronal cells reduced significantly virus yields. However, the inhibitory effect of Roc-A on PHB in NSC-34 cells was not through blocking the CRAF/MEK/ERK pathway as previously reported. Instead, Roc-A treated NSC-34 cells had lower mitochondria-associated PHB and lower ATP levels that correlated with impaired mitochondria integrity. In vivo, EV71-infected mice treated with Roc-A survived longer than the vehicle-treated animals and had significantly lower virus loads in their spinal cord and brain, whereas virus titers in their limb muscles were comparable to controls. Together, this study uncovers PHB as the first host factor that is specifically involved in EV71 neuropathogenesis and a potential drug target to limit neurological complications.
Collapse
Affiliation(s)
- Issac Horng Khit Too
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Isabelle Bonne
- Electron Microscopy Laboratory, Life Sciences Institute, National University of Singapore, Singapore
| | - Eng Lee Tan
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Centre for Biomedical & Life Sciences, Singapore Polytechnic, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sylvie Alonso
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
- * E-mail:
| |
Collapse
|
35
|
Luo F, Liu J, Wang Y, Xu M, Ren Z. PGG impairs herpes simplex virus type 1 infection via blocking capsid assembly. Future Virol 2018. [DOI: 10.2217/fvl-2017-0109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: Pentagalloylglucose (PGG), a hydrolyzable polyphenol was isolated from Phyllanthus emblica, which exhibited a strong inhibitory activity on HSV-1 infection, but its underlying mechanisms have not been completely delineated. Results/methodology: Using TEM, we first observed that PGG blocked the formation and maturation of HSV-1 capsid particles. Hence, we engaged in exploring the molecular mechanisms of PGG on the capsid assembly. At last, we found that PGG also blocked the relocalization of capsid proteins from the cytoplasm to the nucleus where the assembly took place. Conclusion: The current studies, for the first time, demonstrated acetylated microtubules were needed at this process of capsid proteins nuclear translocation. PGG also impairs herpes simplex virus type 1 infection by blocking capsid assembly.
Collapse
Affiliation(s)
- Fan Luo
- Guangzhou Jinan Biomedicine Research & Development Center, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
- The Industry-Academia-Research Demonstration Base of Guangdong Higher Education Institutes (Namely Innovative Culturing Base of Graduates), Jinan University, Guangzhou, Guangdong, China
- College of Life Science & Technology, Jinan University, Guangzhou, Guangdong, China
| | - Junwei Liu
- Guangzhou Jinan Biomedicine Research & Development Center, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yifei Wang
- Guangzhou Jinan Biomedicine Research & Development Center, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
- The Industry-Academia-Research Demonstration Base of Guangdong Higher Education Institutes (Namely Innovative Culturing Base of Graduates), Jinan University, Guangzhou, Guangdong, China
| | - Mingfang Xu
- College of Life Science & Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zhe Ren
- Guangzhou Jinan Biomedicine Research & Development Center, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
- The Industry-Academia-Research Demonstration Base of Guangdong Higher Education Institutes (Namely Innovative Culturing Base of Graduates), Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Len ACL, Starling S, Shivkumar M, Jolly C. HIV-1 Activates T Cell Signaling Independently of Antigen to Drive Viral Spread. Cell Rep 2017; 18:1062-1074. [PMID: 28122231 PMCID: PMC5289937 DOI: 10.1016/j.celrep.2016.12.057] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/28/2016] [Accepted: 12/16/2016] [Indexed: 11/24/2022] Open
Abstract
HIV-1 spreads between CD4 T cells most efficiently through virus-induced cell-cell contacts. To test whether this process potentiates viral spread by activating signaling pathways, we developed an approach to analyze the phosphoproteome in infected and uninfected mixed-population T cells using differential metabolic labeling and mass spectrometry. We discovered HIV-1-induced activation of signaling networks during viral spread encompassing over 200 cellular proteins. Strikingly, pathways downstream of the T cell receptor were the most significantly activated, despite the absence of canonical antigen-dependent stimulation. The importance of this pathway was demonstrated by the depletion of proteins, and we show that HIV-1 Env-mediated cell-cell contact, the T cell receptor, and the Src kinase Lck were essential for signaling-dependent enhancement of viral dissemination. This study demonstrates that manipulation of signaling at immune cell contacts by HIV-1 is essential for promoting virus replication and defines a paradigm for antigen-independent T cell signaling. Unbiased global analysis of T cell signaling changes during HIV-1 cell-cell spread Experimental system to map dynamic signaling changes in mixed cell populations over time More than 200 host cell proteins are modified as HIV-1 disseminates between T cells HIV-1 activates antigen-independent TCR signaling to drive viral spread
Collapse
Affiliation(s)
- Alice C L Len
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Shimona Starling
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Maitreyi Shivkumar
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Clare Jolly
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK.
| |
Collapse
|
37
|
Mitochondrial protein p32/HAPB1/gC1qR/C1qbp is required for efficient respiratory syncytial virus production. Biochem Biophys Res Commun 2017; 489:460-465. [DOI: 10.1016/j.bbrc.2017.05.171] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 05/29/2017] [Indexed: 11/21/2022]
|
38
|
Titanji BK, Pillay D, Jolly C. Combination antiretroviral therapy and cell-cell spread of wild-type and drug-resistant human immunodeficiency virus-1. J Gen Virol 2017; 98:821-834. [PMID: 28141491 PMCID: PMC5657029 DOI: 10.1099/jgv.0.000728] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/27/2017] [Indexed: 12/24/2022] Open
Abstract
Human immunodeficiency virus-1 (HIV-1) disseminates between T cells either by cell-free infection or by highly efficient direct cell-cell spread. The high local multiplicity that characterizes cell-cell infection causes variability in the effectiveness of antiretroviral drugs applied as single agents. Whereas protease inhibitors (PIs) are effective inhibitors of HIV-1 cell-cell and cell-free infection, some reverse transcriptase inhibitors (RTIs) show reduced potency; however, antiretrovirals are not administered as single agents and are used clinically as combination antiretroviral therapy (cART). Here we explored the efficacy of PI- and RTI-based cART against cell-cell spread of wild-type and drug-resistant HIV-1 strains. Using a quantitative assay to measure cell-cell spread of HIV-1 between T cells, we evaluated the efficacy of different clinically relevant drug combinations. We show that combining PIs and RTIs improves the potency of inhibition of HIV-1 and effectively blocks both cell-free and cell-cell spread. Combining drugs that alone are poor inhibitors of cell-cell spread markedly improves HIV-1 inhibition, demonstrating that clinically relevant combinations of ART can inhibit this mode of HIV-1 spread. Furthermore, comparison of wild-type and drug-resistant viruses reveals that PI- and RTI-resistant viruses have a replicative advantage over wild-type virus when spreading by cell-cell means in the presence of cART, suggesting that in the context of inadequate drug combinations or drug resistance, cell-cell spread could potentially allow for ongoing viral replication.
Collapse
Affiliation(s)
- Boghuma Kabisen Titanji
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
- Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Deenan Pillay
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
- Africa Centre for Health and Population Sciences, University of KwaZulu-Natal, KwaZulu-Natal, South Africa
| | - Clare Jolly
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| |
Collapse
|
39
|
Comparative Subcellular Proteomics Analysis of Susceptible and Near-isogenic Resistant Bombyx mori (Lepidoptera) Larval Midgut Response to BmNPV infection. Sci Rep 2017; 7:45690. [PMID: 28361957 PMCID: PMC5374506 DOI: 10.1038/srep45690] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 03/03/2017] [Indexed: 02/01/2023] Open
Abstract
The molecular mechanism of silkworm resistance to Bombyx mori nucleopolyhedrovirus (BmNPV) infection remains largely unclear. Accumulating evidence suggests that subcellular fractionation combined with proteomics is an ideal technique to analyse host antiviral mechanisms. To clarify the anti-BmNPV mechanism of the silkworm, the near-isogenic line BC9 (resistant strain) and the recurrent parent P50 (susceptible strain) were used in a comparative subcellular proteomics study. Two-dimensional gel electrophoresis (2-DE) combined with mass spectrometry (MS) was conducted on proteins extracted from the cytosol, mitochondria, and microsomes of BmNPV-infected and control larval midguts. A total of 87 proteins were successfully identified from the three subcellular fractions. These proteins were primarily involved in energy metabolism, protein metabolism, signalling pathways, disease, and transport. In particular, disease-relevant proteins were especially changed in microsomes. After infection with BmNPV, differentially expressed proteins (DEPs) primarily appeared in the cytosolic and microsomal fractions, which indicated that these two fractions might play a more important role in the response to BmNPV infection. After removing genetic background and individual immune stress response proteins, 16 proteins were identified as potentially involved in repressing BmNPV infection. Of these proteins, the differential expression patterns of 8 proteins according to reverse transcription quantitative PCR (RT-qPCR) analyses were consistent with the 2-DE results.
Collapse
|
40
|
Visualization of HIV T Cell Virological Synapses and Virus-Containing Compartments by Three-Dimensional Correlative Light and Electron Microscopy. J Virol 2017; 91:JVI.01605-16. [PMID: 27847357 PMCID: PMC5215336 DOI: 10.1128/jvi.01605-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/31/2016] [Indexed: 11/20/2022] Open
Abstract
Virological synapses (VS) are adhesive structures that form between infected and uninfected cells to enhance the spread of HIV-1. During T cell VS formation, viral proteins are actively recruited to the site of cell-cell contact where the viral material is efficiently translocated to target cells into heterogeneous, protease-resistant, antibody-inaccessible compartments. Using correlative light and electron microscopy (CLEM), we define the membrane topography of the virus-containing compartments (VCC) where HIV is found following VS-mediated transfer. Focused ion beam scanning electron microscopy (FIB-SEM) and serial sectioning transmission electron microscopy (SS-TEM) were used to better resolve the fluorescent Gag-containing structures within the VCC. We found that small punctate fluorescent signals correlated with single viral particles in enclosed vesicular compartments or surface-localized virus particles and that large fluorescent signals correlated with membranous Gag-containing structures with unknown pathological function. CLEM imaging revealed distinct pools of newly deposited viral proteins within endocytic and nonendocytic compartments in VS target T cells. IMPORTANCE This study directly correlates individual virus-associated objects observed in light microscopy with ultrastructural features seen by electron microscopy in the HIV-1 virological synapse. This approach elucidates which infection-associated ultrastructural features represent bona fide HIV protein complexes. We define the morphology of some HIV cell-to-cell transfer intermediates as true endocytic compartments and resolve unique synapse-associated viral structures created by transfer across virological synapses.
Collapse
|
41
|
Starling S, Jolly C. LFA-1 Engagement Triggers T Cell Polarization at the HIV-1 Virological Synapse. J Virol 2016; 90:9841-9854. [PMID: 27558417 PMCID: PMC5068534 DOI: 10.1128/jvi.01152-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/15/2016] [Indexed: 12/01/2022] Open
Abstract
HIV-1 efficiently disseminates by cell-cell spread at intercellular contacts called virological synapses (VS), where the virus preferentially assembles and buds. Cell-cell contact triggers active polarization of organelles and viral proteins within infected cells to the contact site to support efficient VS formation and HIV-1 spread; critically, however, which cell surface protein triggers contact-induced polarization at the VS remains unclear. Additionally, the mechanism by which the HIV-1 envelope glycoprotein (Env) is recruited to the VS remains ill defined. Here, we use a reductionist bead-coupled antibody assay as a model of the VS and show that cross-linking the integrin LFA-1 alone is sufficient to induce active T cell polarization and recruitment of the microtubule organizing center (MTOC) in HIV-1-infected cells. Mutant cell lines coupled with inhibitors demonstrated that LFA-1-induced polarization was dependent on the T cell kinase ZAP70. Notably, immunofluorescent staining of viral proteins revealed an accumulation of surface Env at sites of LFA-1 engagement, with intracellular Env localized to a Golgi compartment proximal to the polarized MTOC. Furthermore, blocking LFA-1-induced MTOC polarization through ZAP70 inhibition prevented intracellular Env polarization. Taken together, these data reveal that LFA-1 is a key determinant in inducing dynamic T cell remodeling to the VS and suggest a model in which LFA-1 engagement triggers active polarization of the MTOC and the associated Env-containing secretory apparatus to sites of cell-cell contact to support polarized viral assembly and egress for efficient cell-cell spread. IMPORTANCE HIV-1 causes AIDS by spreading within immune cells and depletion of CD4 T lymphocytes. Rapid spread between these cells occurs by highly efficient cell-cell transmission that takes place at virological synapses (VS). VS are characterized by striking T cell remodeling that is spatially associated with polarized virus assembly and budding at sites of cell contact. Here, we show that the integrin LFA-1 triggers organelle polarization and viral protein recruitment, facilitating formation of the VS, and that this requires the T cell kinase ZAP70. Taken together, these data suggest a mechanism by which HIV-1-infected T cells sense and respond to cell contact to polarize viral egress and promote cell-cell spread. Understanding how cell-cell spread is regulated may help reveal therapeutic targets to specifically block this mode of HIV-1 dissemination.
Collapse
Affiliation(s)
- Shimona Starling
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Clare Jolly
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
42
|
HIV-1 Gag, Envelope, and Extracellular Determinants Cooperate To Regulate the Stability and Turnover of Virological Synapses. J Virol 2016; 90:6583-6597. [PMID: 27170746 DOI: 10.1128/jvi.00600-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/02/2016] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Retroviruses spread more efficiently when infected and uninfected cells form tight, physical interfaces known as virological synapses (VSs). VS formation is initiated by adhesive interactions between viral Envelope (Env) glycoproteins on the infected cell and CD4 receptor molecules on the uninfected cell. How high-avidity Env-CD4 linkages are resolved over time is unknown. We describe here a tractable two-color, long-term (>24 h) live cell imaging strategy to study VS turnover in the context of a large cell population, quantitatively. We show that Env's conserved cytoplasmic tail (CT) can potently signal the recruitment of Gag capsid proteins to the VS, a process also dependent on residues within Gag's N-terminal matrix (MA) domain. Additionally, we demonstrate that Env's CT and Gag's MA domain both regulate the duration of interactions between viral donor and target cells, as well as the stability of this interaction over time (i.e., its capacity to resolve or form a syncytium). Finally, we report the unexpected finding that modulating extracellular fluid viscosity markedly impacts target T cell trafficking and thus affects the duration, stability, and turnover of virus-induced cell-cell contacts. Combined, these results suggest a stepwise model for viral cell-to-cell transmission wherein (i) Env-receptor interactions anchor target cells to infected cells, (ii) Env signals Gag's recruitment to the cell-cell contact dependent on an intact Env CT and Gag MA, and (iii) Env CT and Gag MA, in conjunction with extracellular forces, combine to regulate VS stability and infectious outcomes. IMPORTANCE HIV-1 spreads efficiently at physical, cell-cell interfaces known as virological synapses (VSs). The VS provides for spatiotemporal coupling of virus assembly and entry into new host cells and may transmit signals relevant to pathogenesis. Disrupting this mode of transmission may be critical to the goal of abolishing viral persistence in infected individuals. We describe here a long-term live cell imaging strategy for studying virus-induced effects on cell behavior in the context of a large cell population. We demonstrate cooperative roles for viral Gag capsid proteins and Envelope glycoproteins in regulating VS formation and turnover. We also show that modulating fluid viscosity markedly affects T cell trafficking and VS stability. Thus, extracellular factors also play an important role in modulating the nature of infectious cell-cell interactions. In sum, our study provides new tools and insights relevant to exposing vulnerabilities in how HIV-1 and other viruses spread infection among cells, tissues, and people.
Collapse
|
43
|
Identification of Vimentin as a Potential Therapeutic Target against HIV Infection. Viruses 2016; 8:v8060098. [PMID: 27314381 PMCID: PMC4926169 DOI: 10.3390/v8060098] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 04/01/2016] [Accepted: 04/06/2016] [Indexed: 12/22/2022] Open
Abstract
A combination of antiviral drugs known as antiretroviral therapy (ART) has shown effectiveness against the human immunodeficiency virus (HIV). ART has markedly decreased mortality and morbidity among HIV-infected patients, having even reduced HIV transmission. However, an important current disadvantage, resistance development, remains to be solved. Hope is focused on developing drugs against cellular targets. This strategy is expected to prevent the emergence of viral resistance. In this study, using a comparative proteomic approach in MT4 cells treated with an anti-HIV leukocyte extract, we identified vimentin, a molecule forming intermediate filaments in the cell, as a possible target against HIV infection. We demonstrated a strong reduction of an HIV-1 based lentivirus expressing the enhanced green fluorescent protein (eGFP) in vimentin knockdown cells, and a noteworthy decrease of HIV-1 capsid protein antigen (CAp24) in those cells using a multiround infectivity assay. Electron micrographs showed changes in the structure of intermediate filaments when MT4 cells were treated with an anti-HIV leukocyte extract. Changes in the structure of intermediate filaments were also observed in vimentin knockdown MT4 cells. A synthetic peptide derived from a cytoskeleton protein showed potent inhibitory activity on HIV-1 infection, and low cytotoxicity. Our data suggest that vimentin can be a suitable target to inhibit HIV-1.
Collapse
|
44
|
Viruses exploit the tissue physiology of the host to spread in vivo. Curr Opin Cell Biol 2016; 41:81-90. [PMID: 27149407 DOI: 10.1016/j.ceb.2016.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/11/2016] [Accepted: 04/20/2016] [Indexed: 02/07/2023]
Abstract
Viruses are pathogens that strictly depend on their host for propagation. Over years of co-evolution viruses have become experts in exploiting the host cell biology and physiology to ensure efficient replication and spread. Here, we will first summarize the concepts that have emerged from in vitro cell culture studies to understand virus spread. We will then review the results from studies in living animals that reveal how viruses exploit the natural flow of body fluids, specific tissue architecture, and patterns of cell circulation and migration to spread within the host. Understanding tissue physiology will be critical for the design of antiviral strategies that prevent virus dissemination.
Collapse
|
45
|
Abstract
Human immunodeficiency virus type 1 (HIV-1) gives rise to a chronic infection that progressively depletes CD4(+) T lymphocytes. CD4(+) T lymphocytes play a central coordinating role in adaptive cellular and humoral immune responses, and to do so they migrate and interact within lymphoid compartments and at effector sites to mount immune responses. While cell-free virus serves as an excellent prognostic indicator for patient survival, interactions of infected T cells or virus-scavenging immune cells with uninfected T cells can greatly enhance viral spread. HIV can induce interactions between infected and uninfected T cells that are triggered by cell surface expression of viral Env, which serves as a cell adhesion molecule that interacts with CD4 on the target cell, before it acts as the viral membrane fusion protein. These interactions are called virological synapses and promote replication in the face of selective pressure of humoral immune responses and antiretroviral therapy. Other infection-enhancing cell-cell interactions occur between virus-concentrating antigen-presenting cells and recipient T cells, called infectious synapses. The exact roles that these cell-cell interactions play in each stage of infection, from viral acquisition, systemic dissemination, to chronic persistence are still being determined. Infection-promoting immune cell interactions are likely to contribute to viral persistence and enhance the ability of HIV-1 to evade adaptive immune responses.
Collapse
Affiliation(s)
- K M Law
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - N Satija
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - A M Esposito
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - B K Chen
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
46
|
de Armas-Rillo L, Valera MS, Marrero-Hernández S, Valenzuela-Fernández A. Membrane dynamics associated with viral infection. Rev Med Virol 2016; 26:146-60. [PMID: 26817660 PMCID: PMC5066672 DOI: 10.1002/rmv.1872] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 12/15/2022]
Abstract
Viral replication and spreading are fundamental events in the viral life cycle, accounting for the assembly and egression of nascent virions, events that are directly associated with viral pathogenesis in target hosts. These processes occur in cellular compartments that are modified by specialized viral proteins, causing a rearrangement of different cell membranes in infected cells and affecting the ER, mitochondria, Golgi apparatus, vesicles and endosomes, as well as processes such as autophagic membrane flux. In fact, the activation or inhibition of membrane trafficking and other related activities are fundamental to ensure the adequate replication and spreading of certain viruses. In this review, data will be presented that support the key role of membrane dynamics in the viral cycle, especially in terms of the assembly, egression and infection processes. By defining how viruses orchestrate these events it will be possible to understand how they successfully complete their route of infection, establishing viral pathogenesis and provoking disease. © 2015 The Authors Reviews in Medical Virology Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Laura de Armas-Rillo
- Laboratorio de Inmunología Celular y Viral, Unidad de Virología IUETSPC, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), Tenerife, Spain
| | - María-Soledad Valera
- Laboratorio de Inmunología Celular y Viral, Unidad de Virología IUETSPC, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), Tenerife, Spain
| | - Sara Marrero-Hernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Virología IUETSPC, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), Tenerife, Spain
| | - Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Virología IUETSPC, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), Tenerife, Spain
| |
Collapse
|
47
|
A high throughput Cre-lox activated viral membrane fusion assay identifies pharmacological inhibitors of HIV entry. Virology 2016; 490:6-16. [PMID: 26803470 DOI: 10.1016/j.virol.2015.10.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/13/2022]
Abstract
Enveloped virus entry occurs when viral and cellular membranes fuse releasing particle contents into the target cell. Human immunodeficiency virus (HIV) entry occurs by cell-free virus or virus transferred between infected and uninfected cells through structures called virological synapses. We developed a high-throughput cell-based assay to identify small molecule inhibitors of cell-free or virological synapse-mediated entry. An HIV clone carrying Cre recombinase as a Gag-internal gene fusion releases active Cre into cells upon viral entry activating a recombinatorial gene switch changing dsRed to GFP-expression. A screen of a 1998 known-biological profile small molecule library identified pharmacological HIV entry inhibitors that block both cell-free and cell-to-cell infection. Many top hits were noted as HIV inhibitors in prior studies, but not previously recognized as entry antagonists. Modest therapeutic indices for simvastatin and nigericin were observed in confirmatory HIV infection assays. This robust assay is adaptable to study HIV and heterologous viral pseudotypes.
Collapse
|
48
|
Bao Q, Hipp M, Hugo A, Lei J, Liu Y, Kehl T, Hechler T, Löchelt M. In Vitro Evolution of Bovine Foamy Virus Variants with Enhanced Cell-Free Virus Titers and Transmission. Viruses 2015; 7:5855-74. [PMID: 26569290 PMCID: PMC4664980 DOI: 10.3390/v7112907] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 11/16/2022] Open
Abstract
Virus transmission is essential for spreading viral infections and is a highly coordinated process which occurs by cell-free transmission or cell-cell contact. The transmission of Bovine Foamy Virus (BFV) is highly cell-associated, with undetectable cell-free transmission. However, BFV particle budding can be induced by overexpression of wild-type (wt) BFV Gag and Env or artificial retargeting of Gag to the plasma membrane via myristoylation membrane targeting signals, closely resembling observations in other foamy viruses. Thus, the particle release machinery of wt BFV appears to be an excellent model system to study viral adaption to cell-free transmission by in vitro selection and evolution. Using selection for BFV variants with high cell-free infectivity in bovine and non-bovine cells, infectivity dramatically increased from almost no infectious units to about 105-106 FFU (fluorescent focus forming units)/mL in both cell types. Importantly, the selected BFV variants with high titer (HT) cell-free infectivity could still transmit via cell-cell contacts and were neutralized by serum from naturally infected cows. These selected HT-BFV variants will shed light into virus transmission and potential routes of intervention in the spread of viral infections. It will also allow the improvement or development of new promising approaches for antiretroviral therapies.
Collapse
Affiliation(s)
- Qiuying Bao
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
| | - Michaela Hipp
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
| | - Annette Hugo
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
| | - Janet Lei
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK.
| | - Yang Liu
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
- Department Viral Recombination, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA.
| | - Timo Kehl
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
| | - Torsten Hechler
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
- Heidelberg Pharma GmbH, 68526 Ladenburg, Germany.
| | - Martin Löchelt
- Division of Molecuar Diagnostics of Oncogenic Infections, Research Focus Infection, Inflammation and Cancer, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Im Neuenheimer Feld 242, 69120, Germany.
| |
Collapse
|
49
|
Jeng MY, Ali I, Ott M. Manipulation of the host protein acetylation network by human immunodeficiency virus type 1. Crit Rev Biochem Mol Biol 2015; 50:314-25. [PMID: 26329395 PMCID: PMC4816045 DOI: 10.3109/10409238.2015.1061973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Over the past 15 years, protein acetylation has emerged as a globally important post-translational modification that fine-tunes major cellular processes in many life forms. This dynamic regulatory system is critical both for complex eukaryotic cells and for the viruses that infect them. HIV-1 accesses the host acetylation network by interacting with several key enzymes, thereby promoting infection at multiple steps during the viral life cycle. Inhibitors of host histone deacetylases and bromodomain-containing proteins are now being pursued as therapeutic strategies to enhance current antiretroviral treatment. As more acetylation-targeting compounds are reaching clinical trials, it is time to review the role of reversible protein acetylation in HIV-infected CD4(+) T cells.
Collapse
Affiliation(s)
- Mark Y. Jeng
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Ibraheem Ali
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Melanie Ott
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
50
|
HIV-1 Cell-Free and Cell-to-Cell Infections Are Differentially Regulated by Distinct Determinants in the Env gp41 Cytoplasmic Tail. J Virol 2015; 89:9324-37. [PMID: 26136566 DOI: 10.1128/jvi.00655-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/23/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The HIV-1 envelope (Env) glycoprotein mediates viral entry during both cell-free and cell-to-cell infection of CD4(+) T cells. The highly conserved long cytoplasmic tail (CT) of Env is required in a cell type-dependent manner for optimal infectivity of cell-free virus. To probe the role of the CT in cell-to-cell infection, we tested a panel of mutations in the CT region that maintain or attenuate cell-free infection to investigate whether the functions of the CT are conserved during cell-free and cell-to-cell infection. The mutations tested included truncations of structural motifs in the gp41 CT and two point mutations in lentiviral lytic peptide 3 (LLP-3) previously described as disrupting the infectivity of cell-free virus. We found that small truncations of 28 to 43 amino acids (aa) or two LLP-3 point mutations, YW_SL and LL_RQ, severely impaired single-round cell-free infectivity 10-fold or more relative to wild-type full-length CT. These mutants showed a modest 2-fold reduction in cell-to-cell infection assays. Conversely, large truncations of 93 to 124 aa severely impaired cell-to-cell infectivity 20-fold or more while resulting in a 50% increase in infectivity of cell-free viral particles when produced in 293T cells. Intermediate truncations of 46 to 90 aa showed profound impairment of both modes of infection. Our results show that the abilities of Env to support cell-free and cell-to-cell infection are genetically distinct. These differences are cell type dependent for large-CT-truncation mutants. Additionally, point mutants in LLP-3 can maintain multiround propagation from cell-to-cell in primary CD4(+) T cells. IMPORTANCE The functions of HIV Env gp41 CT remain poorly understood despite being widely studied in the context of cell-free infection. We have identified domains of the gp41 CT responsible for striking selective deficiencies in either cell-free or cell-to-cell infectivity. These differences may reflect a different intrinsic regulatory influence of the CT on cell-associated versus particle-associated Env or differential interaction with host or viral proteins. Our findings provide novel insight into the key regulatory potential of the gp41 CT in cell-free and cell-to-cell HIV-1 infection, particularly for short-truncation mutants of ≤43 amino acids or mutants with point mutations in the LLP-3 helical domain of the CT, which are able to propagate via cell-to-cell infection in the absence of infectious cell-free virus production. These mutants may also serve as tools to further define the contributions of cell-free and cell-to-cell infection in vitro and in vivo.
Collapse
|