1
|
Gerg A, Dobrovolny HM. Quantifying Impact of HIV Receptor Surface Density Reveals Differences in Fusion Dynamics of HIV Strains. Viruses 2025; 17:583. [PMID: 40285025 PMCID: PMC12031222 DOI: 10.3390/v17040583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Human Immunodeficiency Virus (HIV) Type-1 has been studied heavily for decades, yet one area that is still poorly understood is the virus' ability to cause cell-cell fusion. In HIV, the fusion process is mediated by viral surface glycoproteins that bind to CD4 cell receptors. This virus-mediated cell fusion creates multi-nucleated cells called syncytia that can affect infection dynamics. Syncytia formation is often studied using a cell-cell fusion assay, in which donor cells expressing the viral surface protein fuse with acceptor cells expressing the cell receptor. A mathematical model capable of reproducing the dynamics of the cell-cell fusion assay was recently developed and can be used to quantify changes in syncytia formation. In this study, we use this mathematical model to quantify the changes in syncytia formation in HIV as the surface density of the glycoproteins is varied. We find that we need to modify the model to explicitly include a density-dependent syncytia formation rate that allows us to capture the dynamics of the cell-cell fusion assay as the density of the glycoproteins changes. With this modification, we find that cell-cell fusion of the HXB2 strain, which uses the CXCR4 coreceptor, shows a threshold-like behavior, while cell-cell fusion of the Sf162 strain, which uses the CCR5 co-receptor, shows a more gradual change as surface density decreases.
Collapse
Affiliation(s)
| | - Hana M. Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX 76129, USA;
| |
Collapse
|
2
|
Lin MH, Hu LJ, Miller JS, Huang XJ, Zhao XY. CAR-NK cell therapy: a potential antiviral platform. Sci Bull (Beijing) 2025; 70:765-777. [PMID: 39837721 DOI: 10.1016/j.scib.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/31/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Viral infections persist as a significant cause of morbidity and mortality worldwide. Conventional therapeutic approaches often fall short in fully eliminating viral infections, primarily due to the emergence of drug resistance. Natural killer (NK) cells, one of the important members of the innate immune system, possess potent immunosurveillance and cytotoxic functions, thereby playing a crucial role in the host's defense against viral infections. Chimeric antigen receptor (CAR)-NK cell therapy has been developed to redirect the cytotoxic function of NK cells specifically towards virus-infected cells, further enhancing their cytotoxic efficacy. In this manuscript, we review the role of NK cells in antiviral infections and explore the mechanisms by which viruses evade immune detection. Subsequently, we focus on the optimization strategies for CAR-NK cell therapy to address existing limitations. Furthermore, we discuss significant advancements in CAR-NK cell therapy targeting viral infections, including those caused by severe acute respiratory syndrome coronavirus 2, human immunodeficiency virus, hepatitis B virus, human cytomegalovirus, and Epstein-Barr virus.
Collapse
Affiliation(s)
- Ming-Hao Lin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China
| | - Li-Juan Hu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, 55455, USA.
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China; Peking-Tsinghua Center for Life Sciences, Beijing 100871, China.
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing 100044, China.
| |
Collapse
|
3
|
Heil M. Self-DNA driven inflammation in COVID-19 and after mRNA-based vaccination: lessons for non-COVID-19 pathologies. Front Immunol 2024; 14:1259879. [PMID: 38439942 PMCID: PMC10910434 DOI: 10.3389/fimmu.2023.1259879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/26/2023] [Indexed: 03/06/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic triggered an unprecedented concentration of economic and research efforts to generate knowledge at unequalled speed on deregulated interferon type I signalling and nuclear factor kappa light chain enhancer in B-cells (NF-κB)-driven interleukin (IL)-1β, IL-6, IL-18 secretion causing cytokine storms. The translation of the knowledge on how the resulting systemic inflammation can lead to life-threatening complications into novel treatments and vaccine technologies is underway. Nevertheless, previously existing knowledge on the role of cytoplasmatic or circulating self-DNA as a pro-inflammatory damage-associated molecular pattern (DAMP) was largely ignored. Pathologies reported 'de novo' for patients infected with Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV)-2 to be outcomes of self-DNA-driven inflammation in fact had been linked earlier to self-DNA in different contexts, e.g., the infection with Human Immunodeficiency Virus (HIV)-1, sterile inflammation, and autoimmune diseases. I highlight particularly how synergies with other DAMPs can render immunogenic properties to normally non-immunogenic extracellular self-DNA, and I discuss the shared features of the gp41 unit of the HIV-1 envelope protein and the SARS-CoV 2 Spike protein that enable HIV-1 and SARS-CoV-2 to interact with cell or nuclear membranes, trigger syncytia formation, inflict damage to their host's DNA, and trigger inflammation - likely for their own benefit. These similarities motivate speculations that similar mechanisms to those driven by gp41 can explain how inflammatory self-DNA contributes to some of most frequent adverse events after vaccination with the BNT162b2 mRNA (Pfizer/BioNTech) or the mRNA-1273 (Moderna) vaccine, i.e., myocarditis, herpes zoster, rheumatoid arthritis, autoimmune nephritis or hepatitis, new-onset systemic lupus erythematosus, and flare-ups of psoriasis or lupus. The hope is to motivate a wider application of the lessons learned from the experiences with COVID-19 and the new mRNA vaccines to combat future non-COVID-19 diseases.
Collapse
Affiliation(s)
- Martin Heil
- Departamento de Ingeniería Genética, Laboratorio de Ecología de Plantas, Centro de Investigación y de Estudios Avanzados (CINVESTAV)-Unidad Irapuato, Irapuato, Mexico
| |
Collapse
|
4
|
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
|
5
|
Beckford-Vera DR, Flavell RR, Seo Y, Martinez-Ortiz E, Aslam M, Thanh C, Fehrman E, Pardons M, Kumar S, Deitchman AN, Ravanfar V, Schulte B, Wu IWK, Pan T, Reeves JD, Nixon CC, Iyer NS, Torres L, Munter SE, Hyunh T, Petropoulos CJ, Hoh R, Franc BL, Gama L, Koup RA, Mascola JR, Chomont N, Deeks SG, VanBrocklin HF, Henrich TJ. First-in-human immunoPET imaging of HIV-1 infection using 89Zr-labeled VRC01 broadly neutralizing antibody. Nat Commun 2022; 13:1219. [PMID: 35264559 PMCID: PMC8907355 DOI: 10.1038/s41467-022-28727-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 02/01/2022] [Indexed: 11/09/2022] Open
Abstract
A major obstacle to achieving long-term antiretroviral (ART) free remission or functional cure of HIV infection is the presence of persistently infected cells that establish a long-lived viral reservoir. HIV largely resides in anatomical regions that are inaccessible to routine sampling, however, and non-invasive methods to understand the longitudinal tissue-wide burden of HIV persistence are urgently needed. Positron emission tomography (PET) imaging is a promising strategy to identify and characterize the tissue-wide burden of HIV. Here, we assess the efficacy of using immunoPET imaging to characterize HIV reservoirs and identify anatomical foci of persistent viral transcriptional activity using a radiolabeled HIV Env-specific broadly neutralizing antibody, 89Zr-VRC01, in HIV-infected individuals with detectable viremia and on suppressive ART compared to uninfected controls (NCT03729752). We also assess the relationship between PET tracer uptake in tissues and timing of ART initiation and direct HIV protein expression in CD4 T cells obtained from lymph node biopsies. We observe significant increases in 89Zr-VRC01 uptake in various tissues (including lymph nodes and gut) in HIV-infected individuals with detectable viremia (N = 5) and on suppressive ART (N = 5) compared to uninfected controls (N = 5). Importantly, PET tracer uptake in inguinal lymph nodes in viremic and ART-suppressed participants significantly and positively correlates with HIV protein expression measured directly in tissue. Our strategy may allow non-invasive longitudinal characterization of residual HIV infection and lays the framework for the development of immunoPET imaging in a variety of other infectious diseases.
Collapse
Affiliation(s)
- Denis R Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Enrique Martinez-Ortiz
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Maya Aslam
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Cassandra Thanh
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Emily Fehrman
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Marion Pardons
- Department of Microbiology, Infectiology and Immunology, Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
| | - Shreya Kumar
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Amelia N Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, USA
| | - Vahid Ravanfar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Brailee Schulte
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - I-Wei Katherine Wu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Tony Pan
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jacqueline D Reeves
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christopher C Nixon
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nikita S Iyer
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Leonel Torres
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sadie E Munter
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Tony Hyunh
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Christos J Petropoulos
- Monogram Biosciences, Inc., Laboratory Corporation of America, South San Francisco, San Francisco, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Benjamin L Franc
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Lucio Gama
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicolas Chomont
- Department of Microbiology, Infectiology and Immunology, Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
The endoplasmic reticulum proteostasis network profoundly shapes the protein sequence space accessible to HIV envelope. PLoS Biol 2022; 20:e3001569. [PMID: 35180219 PMCID: PMC8906867 DOI: 10.1371/journal.pbio.3001569] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 03/09/2022] [Accepted: 02/07/2022] [Indexed: 12/27/2022] Open
Abstract
The sequence space accessible to evolving proteins can be enhanced by cellular chaperones that assist biophysically defective clients in navigating complex folding landscapes. It is also possible, at least in theory, for proteostasis mechanisms that promote strict quality control to greatly constrain accessible protein sequence space. Unfortunately, most efforts to understand how proteostasis mechanisms influence evolution rely on artificial inhibition or genetic knockdown of specific chaperones. The few experiments that perturb quality control pathways also generally modulate the levels of only individual quality control factors. Here, we use chemical genetic strategies to tune proteostasis networks via natural stress response pathways that regulate the levels of entire suites of chaperones and quality control mechanisms. Specifically, we upregulate the unfolded protein response (UPR) to test the hypothesis that the host endoplasmic reticulum (ER) proteostasis network shapes the sequence space accessible to human immunodeficiency virus-1 (HIV-1) envelope (Env) protein. Elucidating factors that enhance or constrain Env sequence space is critical because Env evolves extremely rapidly, yielding HIV strains with antibody- and drug-escape mutations. We find that UPR-mediated upregulation of ER proteostasis factors, particularly those controlled by the IRE1-XBP1s UPR arm, globally reduces Env mutational tolerance. Conserved, functionally important Env regions exhibit the largest decreases in mutational tolerance upon XBP1s induction. Our data indicate that this phenomenon likely reflects strict quality control endowed by XBP1s-mediated remodeling of the ER proteostasis environment. Intriguingly, and in contrast, specific regions of Env, including regions targeted by broadly neutralizing antibodies, display enhanced mutational tolerance when XBP1s is induced, hinting at a role for host proteostasis network hijacking in potentiating antibody escape. These observations reveal a key function for proteostasis networks in decreasing instead of expanding the sequence space accessible to client proteins, while also demonstrating that the host ER proteostasis network profoundly shapes the mutational tolerance of Env in ways that could have important consequences for HIV adaptation. The host cell’s endoplasmic reticulum proteostasis network has a profound, constraining impact on the protein sequence space accessible to HIV’s envelope protein, which is a major target of the host’s adaptive immune system; in particular, upregulation of stringent quality control pathways appears to restrict the viability of destabilizing envelope variants.
Collapse
|
7
|
Wang L, Sandmeyer A, Hübner W, Li H, Huser T, Chen BK. A Replication-Competent HIV Clone Carrying GFP-Env Reveals Rapid Env Recycling at the HIV-1 T Cell Virological Synapse. Viruses 2021; 14:v14010038. [PMID: 35062242 PMCID: PMC8781834 DOI: 10.3390/v14010038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/08/2021] [Accepted: 12/17/2021] [Indexed: 12/24/2022] Open
Abstract
HIV-1 infection is enhanced by cell-cell adhesions between infected and uninfected T cells called virological synapses (VS). VS are initiated by the interactions of cell-surface HIV-1 envelope glycoprotein (Env) and CD4 on target cells and act as sites of viral assembly and viral transfer between cells. To study the process that recruits and retains HIV-1 Env at the VS, a replication-competent HIV-1 clone carrying an Env-sfGFP fusion protein was designed to enable live tracking of Env within infected cells. Combined use of surface pulse-labeling of Env and fluorescence recovery after photobleaching (FRAP) studies, enabled the visualization of the targeted accumulation and sustained recycling of Env between endocytic compartments (EC) and the VS. We observed dynamic exchange of Env at the VS, while the viral structural protein, Gag, was largely immobile at the VS. The disparate exchange rates of Gag and Env at the synapse support that the trafficking and/or retention of a majority of Env towards the VS is not maintained by entrapment by a Gag lattice or immobilization by binding to CD4 on the target cell. A FRAP study of an Env endocytosis mutant showed that recycling is not required for accumulation at the VS, but is required for the rapid exchange of Env at the VS. We conclude that the mechanism of Env accumulation at the VS and incorporation into nascent particles involves continuous internalization and targeted secretion rather than irreversible interactions with the budding virus, but that this recycling is largely dispensable for VS formation and viral transfer across the VS.
Collapse
Affiliation(s)
- Lili Wang
- Department of Medicine, Division of Infectious Disease, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (L.W.); (H.L.)
| | - Alice Sandmeyer
- Biomolecular Photonics, Department of Physics, University of Bielefeld, 33615 Bielefeld, Germany; (A.S.); (W.H.); (T.H.)
| | - Wolfgang Hübner
- Biomolecular Photonics, Department of Physics, University of Bielefeld, 33615 Bielefeld, Germany; (A.S.); (W.H.); (T.H.)
| | - Hongru Li
- Department of Medicine, Division of Infectious Disease, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (L.W.); (H.L.)
| | - Thomas Huser
- Biomolecular Photonics, Department of Physics, University of Bielefeld, 33615 Bielefeld, Germany; (A.S.); (W.H.); (T.H.)
| | - Benjamin K. Chen
- Department of Medicine, Division of Infectious Disease, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (L.W.); (H.L.)
- Correspondence:
| |
Collapse
|
8
|
Lim RM, Rong L, Zhen A, Xie J. A Universal CAR-NK Cell Targeting Various Epitopes of HIV-1 gp160. ACS Chem Biol 2020; 15:2299-2310. [PMID: 32667183 DOI: 10.1021/acschembio.0c00537] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Engineering T cells and natural killer (NK) cells with anti-HIV chimeric antigen receptors (CAR) has emerged as a promising strategy to eradicate HIV-infected cells. However, current anti-HIV CARs are limited by targeting a single epitope of the HIV envelope glycoprotein gp160, which cannot counter the enormous diversity and mutability of viruses. Here, we report the development of a universal CAR-NK cell, which recognizes 2,4-dinitrophenyl (DNP) and can subsequently be redirected to target various epitopes of gp160 using DNP-conjugated antibodies as adaptor molecules. We show that this CAR-NK cell can recognize and kill mimic HIV-infected cell lines expressing subtypes B and C gp160. We additionally find that anti-gp160 antibodies targeting membrane-distal epitopes (including V1/V2, V3, and CD4bs) are more likely to activate universal CAR-NK cells against gp160+ target cells, compared with those targeting membrane-proximal epitopes located in the gp41 MPER. Finally, we confirm that HIV-infected primary human CD4+ T cells can be effectively killed using the same approach. Given that numerous anti-gp160 antibodies with different antigen specificities are readily available, this modular universal CAR-NK cell platform can potentially overcome HIV diversity, thus providing a promising strategy to eradicate HIV-infected cells.
Collapse
Affiliation(s)
- Rebecca M. Lim
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, United States
| | - Liang Rong
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, United States
| | - Anjie Zhen
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, California 90095, United States
| | - Jianming Xie
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, United States
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
9
|
Sengupta P, Lippincott-Schwartz J. Revisiting Membrane Microdomains and Phase Separation: A Viral Perspective. Viruses 2020; 12:v12070745. [PMID: 32664429 PMCID: PMC7412473 DOI: 10.3390/v12070745] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
Retroviruses selectively incorporate a specific subset of host cell proteins and lipids into their outer membrane when they bud out from the host plasma membrane. This specialized viral membrane composition is critical for both viral survivability and infectivity. Here, we review recent findings from live cell imaging of single virus assembly demonstrating that proteins and lipids sort into the HIV retroviral membrane by a mechanism of lipid-based phase partitioning. The findings showed that multimerizing HIV Gag at the assembly site creates a liquid-ordered lipid phase enriched in cholesterol and sphingolipids. Proteins with affinity for this specialized lipid environment partition into it, resulting in the selective incorporation of proteins into the nascent viral membrane. Building on this and other work in the field, we propose a model describing how HIV Gag induces phase separation of the viral assembly site through a mechanism involving transbilayer coupling of lipid acyl chains and membrane curvature changes. Similar phase-partitioning pathways in response to multimerizing structural proteins likely help sort proteins into the membranes of other budding structures within cells.
Collapse
|
10
|
Alexandre K, Malatji K, Mulaudzi T. Comparison of the antiviral activity of the microbicide candidate griffithsin and its tandemers derivatives against different modes of HIV-1 transmission. Virology 2020; 544:12-20. [PMID: 32174510 DOI: 10.1016/j.virol.2020.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 01/22/2023]
Abstract
Tandemers 2MG, 2MG3, 3MG and 4MG are derivatives of the potent anti-HIV-1 microbicide candidate griffithsin (GRFT). We compared these compounds anti-HIV-1 activity to GRFT using the viruses CAP206.08 and CAAN5342.A2 that have decreased sensitivity to this lectin. The 2MG and 2MG3 tandemers had similar activity to GRFT against cell-free and cell-associated viruses, while 3MG and 4MG were significantly more potent. Furthermore, the restoration of the 234N or 295N glycan in these viruses, known to increase sensitivity to GRFT, also increased sensitivity to 2MG and 2MG3, and not to 3MG and 4MG. In addition, GRFT resistant viruses generated in-vitro were equally resistant to 2MG and 2MG3 while they had considerably low resistance to 3MG and 4MG. Lastly, all five compounds showed increased inhibitory activity in seminal and vaginal simulants although the effect was more pronounced in the former. These data support further studies of tandemers as potential microbicides.
Collapse
Affiliation(s)
- Kabamba Alexandre
- Council for Scientific and Industrial Research, Emerging Research Area Platform, Next Generation Health Cluster, Pretoria, Gauteng, South Africa.
| | - Kanyane Malatji
- Council for Scientific and Industrial Research, Emerging Research Area Platform, Next Generation Health Cluster, Pretoria, Gauteng, South Africa; University of the Witwatersrand, School of Pathology, Johannesburg, South Africa
| | - Takalani Mulaudzi
- Council for Scientific and Industrial Research, Emerging Research Area Platform, Next Generation Health Cluster, Pretoria, Gauteng, South Africa; University of the Witwatersrand, School of Pathology, Johannesburg, South Africa
| |
Collapse
|
11
|
Sadowski I, Hashemi FB. Strategies to eradicate HIV from infected patients: elimination of latent provirus reservoirs. Cell Mol Life Sci 2019; 76:3583-3600. [PMID: 31129856 PMCID: PMC6697715 DOI: 10.1007/s00018-019-03156-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/29/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023]
Abstract
35 years since identification of HIV as the causative agent of AIDS, and 35 million deaths associated with this disease, significant effort is now directed towards the development of potential cures. Current anti-retroviral (ART) therapies for HIV/AIDS can suppress virus replication to undetectable levels, and infected individuals can live symptom free so long as treatment is maintained. However, removal of therapy allows rapid re-emergence of virus from a highly stable reservoir of latently infected cells that exist as a barrier to elimination of the infection with current ART. Prospects of a cure for HIV infection are significantly encouraged by two serendipitous cases where individuals have entered remission following stem cell transplantation from compatible HIV-resistant donors. However, development of a routine cure that could become available to millions of infected individuals will require a means of specifically purging cells harboring latent HIV, preventing replication of latent provirus, or destruction of provirus genomes by gene editing. Elimination of latently infected cells will require a means of exposing this population, which may involve identification of a natural specific biomarker or therapeutic intervention to force their exposure by reactivation of virus expression. Accordingly, the proposed "Shock and Kill" strategy involves treatment with latency-reversing agents (LRA) to induce HIV provirus expression thus exposing these cells to killing by cellular immunity or apoptosis. Current efforts to enable this strategy are directed at developing improved combinations of LRA to produce broad and robust induction of HIV provirus and enhancing the elimination of cells where replication has been reactivated by targeted immune modulation. Alternative strategies may involve preventing re-emergence virus from latently infected cells by "Lock and Block" intervention, where transcription of provirus is inhibited to prevent virus spread or disruption of the HIV provirus genome by genome editing.
Collapse
Affiliation(s)
- Ivan Sadowski
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Wang L, Izadmehr S, Kamau E, Kong XP, Chen BK. Sequential trafficking of Env and Gag to HIV-1 T cell virological synapses revealed by live imaging. Retrovirology 2019; 16:2. [PMID: 30646921 PMCID: PMC6334456 DOI: 10.1186/s12977-019-0464-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 01/07/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND HIV infection is enhanced by cell adhesions that form between infected and uninfected T cells called virological synapses (VS). VS are initiated by an interaction between Env and CD4 on cell surfaces and result in the recruitment of virus assembly to the site of cell-cell contact. However, the recruitment of Env to the VS and its relationship to Gag recruitment is not well defined. RESULTS To study the trafficking of HIV-1 Env through the VS, we constructed a molecular clone of HIV carrying a green fluorescent protein-Env fusion protein called, HIV Env-isfGFP-∆V1V2. The Env-isfGFP-∆V1V2 fusion protein does not produce virus particles on its own, but can be rescued by cotransfection with full-length HIV constructs and produce virus particles that package the fluorescent Env. These rescued fluorescent Env can participate in VS formation and can be used to directly image CD4-dependent Env transfer across VS from donor to target cells. The movements of fluorescently tagged Gag and Env to the VS and transfer into target cells can be also tracked through live imaging. Time lapse live imaging reveals evidence of limited Env accumulation at the site of cell-cell contact shortly after cell adhesion, followed by Gag re-distribution to contact area. Both Gag and Env can be recruited to form button-like spots characteristic of VS. CONCLUSIONS Env and Gag are recruited to the VS in a coordinated temporal sequence and subsequently transfer together across the synapse into the target cell. Env accumulations, when observed, are earlier than Gag re-distribution to the contact area during formation of VS.
Collapse
Affiliation(s)
- Lili Wang
- 0000 0001 0670 2351grid.59734.3cDepartment of Medicine, Division of Infectious Diseases, Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Sudeh Izadmehr
- 0000 0001 0670 2351grid.59734.3cDepartment of Medicine, Division of Infectious Diseases, Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Edwin Kamau
- 0000 0004 1936 8753grid.137628.9Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 550 First Avenue, New York, NY 10016 USA
| | - Xiang-Peng Kong
- 0000 0004 1936 8753grid.137628.9Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 550 First Avenue, New York, NY 10016 USA
| | - Benjamin K. Chen
- 0000 0001 0670 2351grid.59734.3cDepartment of Medicine, Division of Infectious Diseases, Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029 USA
| |
Collapse
|
13
|
Bastian AR, Ang CG, Kamanna K, Shaheen F, Huang YH, McFadden K, Duffy C, Bailey LD, Sundaram RVK, Chaiken I. Targeting cell surface HIV-1 Env protein to suppress infectious virus formation. Virus Res 2017; 235:33-36. [PMID: 28390972 DOI: 10.1016/j.virusres.2017.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 11/25/2022]
Abstract
HIV-1 Env protein is essential for host cell entry, and targeting Env remains an important antiretroviral strategy. We previously found that a peptide triazole thiol KR13 and its gold nanoparticle conjugate AuNP-KR13 directly and irreversibly inactivate the virus by targeting the Env protein, leading to virus gp120 shedding, membrane disruption and p24 capsid protein release. Here, we examined the consequences of targeting cell-surface Env with the virus inactivators. We found that both agents led to formation of non-infectious virus from transiently transfected HEK293T cells. The budded non-infectious viruses lacked Env gp120 but contained gp41. Importantly, budded virions also retained the capsid protein p24, in stark contrast to p24 leakage from viruses directly treated by these agents and arguing that the agents led to deformed viruses by transforming the cells at a stage before virus budding. We found that the Env inactivators caused gp120 shedding from the transiently transfected HEK293T cells as well as non-producer CHO-K1-gp160 cells. Additionally, AuNP-KR13 was cytotoxic against the virus-producing HEK293T and CHO-K1-gp160 cells, but not untransfected HEK293T or unmodified CHO-K1 cells. The results obtained reinforce the argument that cell-surface HIV-1 Env is metastable, as on virus particles, and provides a conformationally vulnerable target for virus suppression and infectious cell inactivation.
Collapse
Affiliation(s)
- Arangassery Rosemary Bastian
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| | - Charles G Ang
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Kantharaju Kamanna
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Farida Shaheen
- Viral and Molecular Core, Penn Center for AIDS Research, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Yu-Hung Huang
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Karyn McFadden
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Caitlin Duffy
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Lauren D Bailey
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Ramalingam Venkat Kalyana Sundaram
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Irwin Chaiken
- Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, United States.
| |
Collapse
|
14
|
Differences in Env and Gag protein expression patterns and epitope availability in feline immunodeficiency virus infected PBMC compared to infected and transfected feline model cell lines. Virus Res 2017; 227:249-260. [PMID: 27836726 DOI: 10.1016/j.virusres.2016.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/05/2016] [Accepted: 11/05/2016] [Indexed: 11/21/2022]
Abstract
Env and Gag are key components of the FIV virion that are targeted to the plasma membrane for virion assembly. They are both important stimulators and targets of anti-FIV immunity. To investigate and compare the expression pattern and antigenic changes of Gag and Env in various research models, infected PBMC (the natural FIV host cells) and GFox, and transfected CrFK were stained over time with various Env and Gag specific MAbs. In FIV infected GFox and PBMC, Env showed changes in epitope availability for antibody binding during processing and trafficking, which was not seen in transfected CrFK. Interestingly, epitopes exposed on intracellular Env and Env present on the plasma membrane of CrFK and GFox seem to be hidden on plasma membrane expressed Env of FIV infected PBMC. A kinetic follow up of Gag and Env expression showed a polarization of both Gag and Env expression to specific sites at the plasma membrane of PBMC, but not in other cell lines. In conclusion, mature trimeric cell surface expressed Env might be antigenically distinct from intracellular monomeric Env in PBMC and might possibly be unrecognizable by feline humoral immunity. In addition, Env expression is restricted to a small area on the plasma membrane and co-localizes with a large moiety of Gag, which may represent a preferred FIV budding site, or initiation of virological synapses with direct cell-to-cell virus transmission.
Collapse
|
15
|
Nath S, Christian L, Tan SY, Ki S, Ehrlich LIR, Poenie M. Dynein Separately Partners with NDE1 and Dynactin To Orchestrate T Cell Focused Secretion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2090-101. [PMID: 27534551 PMCID: PMC5010990 DOI: 10.4049/jimmunol.1600180] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/14/2016] [Indexed: 11/19/2022]
Abstract
Helper and cytotoxic T cells accomplish focused secretion through the movement of vesicles toward the microtubule organizing center (MTOC) and translocation of the MTOC to the target contact site. In this study, using Jurkat cells and OT-I TCR transgenic primary murine CTLs, we show that the dynein-binding proteins nuclear distribution E homolog 1 (NDE1) and dynactin (as represented by p150(Glued)) form mutually exclusive complexes with dynein, exhibit nonoverlapping distributions in target-stimulated cells, and mediate different transport events. When Jurkat cells expressing a dominant negative form of NDE1 (NDE1-enhanced GFP fusion) were activated by Staphylococcus enterotoxin E-coated Raji cells, NDE1 and dynein failed to accumulate at the immunological synapse (IS) and MTOC translocation was inhibited. Knockdown of NDE1 in Jurkat cells or primary mouse CTLs also inhibited MTOC translocation and CTL-mediated killing. In contrast to NDE1, knockdown of p150(Glued), which depleted the alternative dynein/dynactin complex, resulted in impaired accumulation of CTLA4 and granzyme B-containing intracellular vesicles at the IS, whereas MTOC translocation was not affected. Depletion of p150(Glued) in CTLs also inhibited CTL-mediated lysis. We conclude that the NDE1/Lissencephaly 1 and dynactin complexes separately mediate two key components of T cell-focused secretion, namely translocation of the MTOC and lytic granules to the IS, respectively.
Collapse
Affiliation(s)
- Shubhankar Nath
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| | - Laura Christian
- Department of Molecular Cell and Developmental Biology, University of Texas at Austin, Austin, TX 78712
| | - Sarah Youngsun Tan
- Department of Molecular Cell and Developmental Biology, University of Texas at Austin, Austin, TX 78712
| | - Sanghee Ki
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| | - Martin Poenie
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712; and
| |
Collapse
|
16
|
Lebedev AV, Neshumaev DA, Kazennova EV, Lapovok IA, Laga VY, Tumanov AS, Glushchenko NV, Plotnikova YK, Ponomareva OA, Yarygina EI, Bobkova MR. [Comparative analysis of genetic variants of the HIV-1 circulating in the Irkutsk region in 1999 and 2012]. Vopr Virusol 2016; 61:112-118. [PMID: 36494944 DOI: 10.18821/0507-4088-2016-61-3-112-118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 07/12/2020] [Indexed: 12/13/2022]
Abstract
The pol and env genome regions of the HIV-1 genetic variants circulating in the irkutsk region of russia in 1999 and 2012 were compared. The results of this work showed the dominance of the HIV-1 subtype a IDU-A genetic variant (100%) in this region. No primary resistance mutations in the pol gene in the treatment-naive patients were found. The heterogeneity of the viral population was found to be significantly increased based on the pol and env analysis among HIV-variants isolated in 2012 (12.88% and 2.16%) from the intravenous drug users as compared to HIV-variants that caused the outbreak of the HIV infection in 1999 (1.64% and 0.47%). In addition, the comparison of genetic distances of the pol and env gene sequences in the viruses isolated in 2012 from the HIV-positive persons infected through heterosexual intercourse and intravenous drug use demonstrated that the transmission route influenced the variability of the virus population. Among the viruses of IDU-A variant circulating in the area in 2012 the prevalence of X4-tropic variants was 24.7%.
Collapse
Affiliation(s)
- A V Lebedev
- D.I. Ivanovsky Institute of Virology «Federal Research Centre of Epidemilogy and Microbiology named after the honorary academician N.F. Gamaleya».,Moscow State Academy of Veterinary Medicine and Biotechnology
| | | | - E V Kazennova
- D.I. Ivanovsky Institute of Virology «Federal Research Centre of Epidemilogy and Microbiology named after the honorary academician N.F. Gamaleya»
| | - I A Lapovok
- D.I. Ivanovsky Institute of Virology «Federal Research Centre of Epidemilogy and Microbiology named after the honorary academician N.F. Gamaleya»
| | - V Y Laga
- D.I. Ivanovsky Institute of Virology «Federal Research Centre of Epidemilogy and Microbiology named after the honorary academician N.F. Gamaleya»
| | - A S Tumanov
- D.I. Ivanovsky Institute of Virology «Federal Research Centre of Epidemilogy and Microbiology named after the honorary academician N.F. Gamaleya»
| | - N V Glushchenko
- D.I. Ivanovsky Institute of Virology «Federal Research Centre of Epidemilogy and Microbiology named after the honorary academician N.F. Gamaleya»
| | | | | | - E I Yarygina
- Moscow State Academy of Veterinary Medicine and Biotechnology
| | - M R Bobkova
- D.I. Ivanovsky Institute of Virology «Federal Research Centre of Epidemilogy and Microbiology named after the honorary academician N.F. Gamaleya»
| |
Collapse
|
17
|
Torres-Castro I, Cortés-Rubio CN, Sandoval G, Lamoyi E, Larralde C, Huerta L. Flow cytometry analysis of cell population dynamics and cell cycle during HIV-1 envelope-mediated formation of syncytia in vitro. In Vitro Cell Dev Biol Anim 2014; 50:453-63. [DOI: 10.1007/s11626-013-9724-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/09/2013] [Indexed: 02/01/2023]
|
18
|
Gaudin R, de Alencar BC, Arhel N, Benaroch P. HIV trafficking in host cells: motors wanted! Trends Cell Biol 2013; 23:652-62. [PMID: 24119663 DOI: 10.1016/j.tcb.2013.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 09/05/2013] [Accepted: 09/09/2013] [Indexed: 11/16/2022]
Abstract
Throughout the viral replication cycle, viral proteins, complexes, and particles need to be transported within host cells. These transport events are dependent on the host cell cytoskeleton and molecular motors. However, the mechanisms by which virus is trafficked along cytoskeleton filaments and how molecular motors are recruited and regulated to guarantee successful integration of the viral genome and production of new viruses has only recently begun to be understood. Recent studies on HIV have identified specific molecular motors involved in the trafficking of these viral particles. Here we review recent literature on the transport of HIV components in the cell, provide evidence for the identity and role of molecular motors in this process, and highlight how these trafficking events may be related to those occurring with other viruses.
Collapse
Affiliation(s)
- Raphaël Gaudin
- Institut Curie, Centre de Recherche, 26 rue d'Ulm, 75248 Paris Cedex 05, France; INSERM, U932, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | | | | | | |
Collapse
|
19
|
Retroviral env glycoprotein trafficking and incorporation into virions. Mol Biol Int 2012; 2012:682850. [PMID: 22811910 PMCID: PMC3395148 DOI: 10.1155/2012/682850] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 05/08/2012] [Accepted: 05/31/2012] [Indexed: 11/17/2022] Open
Abstract
Together with the Gag protein, the Env glycoprotein is a major retroviral structural protein and is essential for forming infectious virus particles. Env is synthesized, processed, and transported to certain microdomains at the plasma membrane and takes advantage of the same host machinery for its trafficking as that used by cellular glycoproteins. Incorporation of Env into progeny virions is probably mediated by the interaction between Env and Gag, in some cases with the additional involvement of certain host factors. Although several general models have been proposed to explain the incorporation of retroviral Env glycoproteins into virions, the actual mechanism for this process is still unclear, partly because structural data on the Env protein cytoplasmic tail is lacking. This paper presents the current understanding of the synthesis, trafficking, and virion incorporation of retroviral Env proteins.
Collapse
|
20
|
Jolly C, Welsch S, Michor S, Sattentau QJ. The regulated secretory pathway in CD4(+) T cells contributes to human immunodeficiency virus type-1 cell-to-cell spread at the virological synapse. PLoS Pathog 2011; 7:e1002226. [PMID: 21909273 PMCID: PMC3164651 DOI: 10.1371/journal.ppat.1002226] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 07/05/2011] [Indexed: 11/19/2022] Open
Abstract
Direct cell-cell spread of Human Immunodeficiency Virus type-1 (HIV-1) at the virological synapse (VS) is an efficient mode of dissemination between CD4(+) T cells but the mechanisms by which HIV-1 proteins are directed towards intercellular contacts is unclear. We have used confocal microscopy and electron tomography coupled with functional virology and cell biology of primary CD4(+) T cells from normal individuals and patients with Chediak-Higashi Syndrome and report that the HIV-1 VS displays a regulated secretion phenotype that shares features with polarized secretion at the T cell immunological synapse (IS). Cell-cell contact at the VS re-orientates the microtubule organizing center (MTOC) and organelles within the HIV-1-infected T cell towards the engaged target T cell, concomitant with polarization of viral proteins. Directed secretion of proteins at the T cell IS requires specialized organelles termed secretory lysosomes (SL) and we show that the HIV-1 envelope glycoprotein (Env) localizes with CTLA-4 and FasL in SL-related compartments and at the VS. Finally, CD4(+) T cells that are disabled for regulated secretion are less able to support productive cell-to-cell HIV-1 spread. We propose that HIV-1 hijacks the regulated secretory pathway of CD4(+) T cells to enhance its dissemination.
Collapse
Affiliation(s)
- Clare Jolly
- MRC Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London, United Kingdom.
| | | | | | | |
Collapse
|
21
|
Jolly C. T cell polarization at the virological synapse. Viruses 2010; 2:1261-1278. [PMID: 21994679 PMCID: PMC3185707 DOI: 10.3390/v2061261] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/20/2010] [Accepted: 05/28/2010] [Indexed: 11/16/2022] Open
Abstract
Cell-to-cell spread of HIV-1 between CD4(+) T cells takes place at multimolecular structures called virological synapses. A defining feature of the virological synapse is polarization of viral assembly and budding at sites of T cell-T cell contact. Recent work is beginning to address how viral proteins are targeted to the virological synapse and the molecular mechanisms that regulate HIV-1 egress by cell-to-cell spread. This review discusses our current understanding of these processes and considers how T cell polarization during other forms of intercellular communication may provide insight into HIV-1 assembly and dissemination.
Collapse
Affiliation(s)
- Clare Jolly
- MRC Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, W1T 4JF, UK
| |
Collapse
|
22
|
Vesicle traffic to the immunological synapse: a multifunctional process targeted by lymphotropic viruses. Curr Top Microbiol Immunol 2010; 340:191-207. [PMID: 19960315 DOI: 10.1007/978-3-642-03858-7_10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The site of contact between T lymphocytes and antigen-presenting cells becomes, upon antigen recognition, an organized junction named the immunological synapse. Various T cell organelles polarize, together with microtubules, toward the antigen-presenting cell. Among them, intracellular vesicular compartments, such as the Golgi apparatus, the recycling endosomal compartment, or cytotoxic granules help to build the immunological synapse and ensure effector functions, such as polarized secretion of cytokines by helper T cells, or exocytosis of lytic granules by cytotoxic T cells. Lymphotropic retroviruses, such as the human immunodeficiency virus type 1, the human T cell leukemia virus type 1, or the Herpesvirus saimiri, can subvert some of the vesicle traffic mechanisms impeding the generation and function of the immunological synapses. This review focuses on the polarization of vesicle traffic, its regulation, and its role in maintaining the structure and function of the immunological synapse. We discuss how some lymphotropic viruses target the vesicle traffic in T lymphocytes, inhibiting the formation of immunological synapses and modulating the response of infected T cells.
Collapse
|
23
|
Parker CG, Domaoal RA, Anderson KS, Spiegel DA. An antibody-recruiting small molecule that targets HIV gp120. J Am Chem Soc 2009; 131:16392-4. [PMID: 19839582 PMCID: PMC2783809 DOI: 10.1021/ja9057647] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
HIV/AIDS is a global pandemic for which new treatment strategies are desperately needed. We have designed a novel small molecule, designated as ARM-H, that has the potential to interfere with HIV survival through two mechanisms: (1) by recruiting antibodies to gp120-expressing virus particles and infected human cells, thus enhancing their uptake and destruction by the human immune system, and (2) by binding the viral glycoprotein gp120, inhibiting its interaction with the human protein CD4 and preventing virus entry. Here we demonstrate that ARM-H is capable of simultaneously binding gp120, a component of the Env surface viral glycoprotein (found on the surface of both HIV and virus-infected cells) and anti-2,4-dinitrophenyl antibodies (already present in the human bloodstream). The ternary complex formed between the antibody, ARM-H, and gp120 is immunologically active and leads to the complement-mediated destruction of Env-expressing cells. Furthermore, ARM-H prevents virus entry into human T-cells and should therefore be capable of inhibiting virus replication through two mutually reinforcing mechanisms (inhibition of virus entry and antibody-mediated killing). These studies demonstrate the viable anti-HIV activity of antibody-recruiting small molecules and have the potential to initiate novel paradigms in HIV treatment.
Collapse
Affiliation(s)
- Christopher G. Parker
- Department of Chemistry, Yale University, 225 Prospect Street, PO Box 208107, New Haven, CT 06520-8107
| | - Robert A. Domaoal
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, SHM B350B, New Haven, CT 06520
| | - Karen S. Anderson
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, SHM B350B, New Haven, CT 06520
| | - David A. Spiegel
- Department of Chemistry, Yale University, 225 Prospect Street, PO Box 208107, New Haven, CT 06520-8107
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, SHM B350B, New Haven, CT 06520
| |
Collapse
|
24
|
Haller C, Fackler OT. HIV-1 at the immunological and T-lymphocytic virological synapse. Biol Chem 2009; 389:1253-60. [PMID: 18713012 DOI: 10.1515/bc.2008.143] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cell-cell transmission of human immunodeficiency virus type 1 (HIV-1) is considered the most effective mode of viral spread in T-lymphocyte cultures. Evidence has accumulated that HIV-1 assembles polarized synaptic-like structures, referred to as virological synapses, as specialized sites of viral transfer. Interestingly, it was recently also discovered that HIV-1 impairs the formation of the structurally similar immunological synapse, thereby modulating exogenous T-lymphocyte stimulation to yield an optimal activation state for productive HIV-1 infection. The careful dissection of these opposing effects will contribute to our understanding of retroviral spread and cellular signal transduction machineries.
Collapse
Affiliation(s)
- Claudia Haller
- Department of Virology, University of Heidelberg, INF 324, D-69120 Heidelberg, Germany
| | | |
Collapse
|
25
|
Abstract
Recently, it has been emphasized that chronic generalized immune activation is a leading event in the pathogenesis of HIV-1 infection. Supporting evidence comes from observations that in cases of lack of activation, infected subjects maintain a high number of T cells and do not develop AIDS-related events. Despite intensive studies, the exact mechanisms of T-cell activation are still not well understood and options for their control are limited. Very promising in this direction is a recently described T-cell subpopulation--regulatory T cells. Their functional activity and vitality are strongly dependent on the presence of IL-2. Better understanding of the mechanisms of T-cell activation, as well as the contribution of regulatory T cells to its control will increase therapeutic options for HIV-1-infected subjects. The application of immune-based therapy together with highly active antiretroviral therapy will lend a helping hand to the natural regulatory mechanisms in the control of infection.
Collapse
Affiliation(s)
- Velislava Terzieva
- Department of Immunology, National Center of Infectious and Parasitic Diseases, Sofia, Bulgaria.
| |
Collapse
|
26
|
Perugi F, Muriaux D, Ramirez BC, Chabani S, Decroly E, Darlix JL, Blot V, Pique C. Human Discs Large is a new negative regulator of human immunodeficiency virus-1 infectivity. Mol Biol Cell 2008; 20:498-508. [PMID: 18946087 DOI: 10.1091/mbc.e08-02-0189] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Human immunodeficiency virus (HIV)-1 replication is positively or negatively regulated through multiple interactions with host cell proteins. We report here that human Discs Large (Dlg1), a scaffold protein recruited beneath the plasma membrane and involved in the assembly of multiprotein complexes, restricts HIV-1 infectivity. The endogenous Dlg1 and HIV-1 Gag polyprotein spontaneously interact in HIV-1-chronically infected T cells. Depleting endogenous Dlg1 in either adherent cells or T cells does not affect Gag maturation, production, or release, but it enhances the infectivity of progeny viruses five- to sixfold. Conversely, overexpression of Dlg1 reduces virus infectivity by approximately 80%. Higher virus infectivity upon Dlg1 depletion correlates with increased Env content in cells and virions, whereas the amount of virus-associated Gag or genomic RNA remains identical. Dlg1 knockdown is also associated with the redistribution and colocalization of Gag and Env toward CD63 and CD82 positive vesicle-like structures, including structures that seem to still be connected to the plasma membrane. This study identifies both a new negative regulator that targets the very late steps of the HIV-1 life cycle, and an assembly pathway that optimizes HIV-1 infectivity.
Collapse
Affiliation(s)
- Fabien Perugi
- Department of Cell Biology, Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique Unité Mixte de Recherche, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Murakami T. Roles of the interactions between Env and Gag proteins in the HIV-1 replication cycle. Microbiol Immunol 2008; 52:287-95. [PMID: 18557900 DOI: 10.1111/j.1348-0421.2008.00008.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The Env and Gag proteins of HIV-1 are the two major structural proteins of this retrovirus. The interactions between Env and Gag proteins and their regulation in HIV-1 are required for several steps of the replication cycle, involving not only virus assembly, specifically Env incorporation, but also entry steps after virus maturation. A large number of host factors and certain membrane microdomains appear to engage both in transport/trafficking of Env and/or Gag proteins, and in the interactions of these two proteins. The present review briefly summarizes our current knowledge regarding the roles of the interactions between Env and Gag proteins in the virus replication cycle.
Collapse
|
28
|
Leung K, Kim JO, Ganesh L, Kabat J, Schwartz O, Nabel GJ. HIV-1 assembly: viral glycoproteins segregate quantally to lipid rafts that associate individually with HIV-1 capsids and virions. Cell Host Microbe 2008; 3:285-92. [PMID: 18474355 PMCID: PMC2998762 DOI: 10.1016/j.chom.2008.04.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 04/04/2008] [Accepted: 04/24/2008] [Indexed: 01/08/2023]
Abstract
HIV-1 assembly depends on its structural protein, Gag, which after synthesis on ribosomes, traffics to the late endosome/plasma membrane, associates with HIV Env glycoprotein, and forms infectious virions. While Env and Gag migrate to lipid microdomains, their stoichiometry and specificity of interaction are unknown. Pseudotyped viral particles can be made with one viral core surrounded by heterologous envelope proteins. Taking advantage of this property, we analyzed the association of HIV Env and Ebola glycoprotein (GP), with HIV-1 Gag coexpressed in the same cell. Though both viral glycoproteins were expressed, each associated independently with Gag, giving rise to distinct virion populations, each with a single glycoprotein type. Confocal imaging demonstrated that Env and GP localized to distinct lipid raft microdomains within the same cell where they associated with different virions. Thus, a single Gag particle associates “quantally” with one lipid raft, containing homogeneous trimeric viral envelope proteins, to assemble functional virions.
Collapse
Affiliation(s)
- Kwanyee Leung
- Vaccine Research Center, NIAID, National Institutes of Health, Bethesda, MD 20892-0485, USA
| | | | | | | | | | | |
Collapse
|
29
|
Jolly C, Sattentau QJ. Regulated secretion from CD4+ T cells. Trends Immunol 2007; 28:474-81. [DOI: 10.1016/j.it.2007.08.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 08/16/2007] [Accepted: 08/16/2007] [Indexed: 10/22/2022]
|
30
|
Sol-Foulon N, Sourisseau M, Porrot F, Thoulouze MI, Trouillet C, Nobile C, Blanchet F, di Bartolo V, Noraz N, Taylor N, Alcover A, Hivroz C, Schwartz O. ZAP-70 kinase regulates HIV cell-to-cell spread and virological synapse formation. EMBO J 2007; 26:516-26. [PMID: 17215865 PMCID: PMC1783460 DOI: 10.1038/sj.emboj.7601509] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Accepted: 11/24/2006] [Indexed: 01/29/2023] Open
Abstract
HIV efficiently spreads in lymphocytes, likely through virological synapses (VSs). These cell-cell junctions share some characteristics with immunological synapses, but cellular proteins required for their constitution remain poorly characterized. We have examined here the role of ZAP-70, a key kinase regulating T-cell activation and immunological synapse formation, in HIV replication. In lymphocytes deficient for ZAP-70, or expressing a kinase-dead mutant of the protein, HIV replication was strikingly delayed. We have characterized further this replication defect. ZAP-70 was dispensable for the early steps of viral cycle, from entry to expression of viral proteins. However, in the absence of ZAP-70, intracellular Gag localization was impaired. ZAP-70 was required in infected donor cells for efficient cell-to-cell HIV transmission to recipients and for formation of VSs. These results bring novel insights into the links that exist between T-cell activation and HIV spread, and suggest that HIV usurps components of the immunological synapse machinery to ensure its own spread through cell-to-cell contacts.
Collapse
Affiliation(s)
| | | | - Françoise Porrot
- Groupe Virus et Immunité, Institut Pasteur, CNRS URA1930, France
| | | | - Céline Trouillet
- Groupe Virus et Immunité, Institut Pasteur, CNRS URA1930, France
| | | | - Fabien Blanchet
- Groupe Virus et Immunité, Institut Pasteur, CNRS URA1930, France
| | - Vincenzo di Bartolo
- Unité de Biologie Cellulaire des Lymphocytes, Institut Pasteur, Paris, France
| | - Nelly Noraz
- CNRS UMR5535, Institut de Génétique Moléculaire, Montpellier, France
| | - Naomi Taylor
- CNRS UMR5535, Institut de Génétique Moléculaire, Montpellier, France
| | - Andres Alcover
- Unité de Biologie Cellulaire des Lymphocytes, Institut Pasteur, Paris, France
| | | | - Olivier Schwartz
- Groupe Virus et Immunité, Institut Pasteur, CNRS URA1930, France
- Virus and Immunity Group, Institut Pasteur, CNRS URA 1930, 28 rue du Dr Roux, 75724 Paris Cedex 15, France. Tel.: +33 1 45 68 83 53; fax: +33 1 45 68 89 40; E-mail:
| |
Collapse
|
31
|
Grigorov B, Arcanger F, Roingeard P, Darlix JL, Muriaux D. Assembly of Infectious HIV-1 in Human Epithelial and T-Lymphoblastic Cell Lines. J Mol Biol 2006; 359:848-62. [PMID: 16682056 DOI: 10.1016/j.jmb.2006.04.017] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 03/28/2006] [Accepted: 04/03/2006] [Indexed: 11/25/2022]
Abstract
The canonical view of the ultimate steps of HIV-1 replication is that virus assembly and budding are taking place at the plasma membrane of infected cells. Surprisingly, recent studies revealed that these steps also occur on endosomal membranes in the interior of infected cells, such as macrophages. This prompted us to revisit the site of HIV-1 assembly in human epithelial-like cells and in infected human T-lymphoblastic cells. To address this question, we investigated the intracellular location of the major viral structural components of HIV-1, namely Gag, Env and the genomic RNA. Using a sub-cellular fractionation method, as well as immuno-confocal and electron microscopy, we show that Gag, the Env glycoproteins and the genomic RNA accumulate in late endosomes that contain infectious HIV-1 particles. In epithelial-like 293T cells, HIV-1 assembles and buds both at the plasma membrane and in endosomes, while in chronically infected human T lymphocytes, viral assembly mostly occurs within the cell where large amounts of infectious virions accumulate in endosomal compartments. In addition, HIV-1 release could be enhanced by ionomycin, a drug stimulating calcium-dependent exocytosis. These results favour the view that newly made Gag molecules associate with the genomic RNA in the cytosol, then viral core complexes can be targeted to late endosomes together with Env, where infectious HIV-1 are made and subsequently released by exocytosis.
Collapse
Affiliation(s)
- Boyan Grigorov
- LaboRetro Unité de virologie humaine INSERM U758, IFR128 BioSciences Lyon-Gerland, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69 364 Lyon, France
| | | | | | | | | |
Collapse
|
32
|
Abstract
HIV-1 buds from the surface of activated T lymphocytes. In macrophages, however, newly formed HIV-1 particles amass in the lumen of an intracellular compartment. Here, we demonstrate by live-cell imaging techniques, by immunocytochemistry and by immuno-electron microscopy that HIV-1 structural proteins, particularly the internal structural protein Gag, accumulate at membranes of the late endocytic compartment in a variety of cell types and not just in monocyte/macrophage-derived cells. Recent biochemical and genetic studies have implicated components of the mammalian vacuolar protein sorting pathway in retroviral budding. Together with those observations, our study suggests that HIV-1 morphogenesis is thoroughly rooted in the endosomal system.
Collapse
Affiliation(s)
- Sascha Nydegger
- Department of Microbiology and Molecular Genetics, College of Medicine and CALS, 318 Stafford Hall, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | |
Collapse
|
33
|
Lucas JJ, Domenico J, Gelfand EW. Cyclin-Dependent Kinase 6 Inhibits Proliferation of Human Mammary Epithelial Cells. Mol Cancer Res 2004. [DOI: 10.1158/1541-7786.105.2.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Many defects in cancer cells are in molecules regulating G1-phase cyclin-dependent kinases (cdks), which are responsible for modulating the activities of Rb family growth-suppressing proteins. Models for understanding how such defects affect proliferation assume that cdks are responsible for sequentially phosphorylating, and hence inactivating, the growth-suppressing functions of Rb family proteins, thus promoting cell cycle progression. However, cdks also play a role in formation of growth-suppressing forms of pRb family molecules, including the “hypophosphorylated” species of pRb itself. Here, it is shown that normal human mammary epithelial cells have a high amount of cdk6 protein and activity, but all breast tumor-derived cell lines analyzed had reduced levels, with several having little or no cdk6. Immunohistochemical studies showed reduced levels of cdk6 in breast tumor cells as compared with normal breast tissue in vivo. Cdk6 levels in two breast tumor cell lines were restored to those characteristic of normal human mammary epithelial cells by DNA transfection. The cells had a reduced growth rate compared with parental tumor cells; cells that lost ectopic expression of cdk6 reverted to the faster growth rate of parental cells. Cell lines with restored cdk6 levels accumulated higher amounts of the Rb family protein p130 as well as E2F4, a suppressing member of the E2F family of transcription factors, in their nuclei. The results suggest that cdk6 restrains rather than stimulates breast epithelial cell proliferation and that its loss or down-regulation could play a role in breast tumor development.
Collapse
Affiliation(s)
- Joseph J. Lucas
- Division of Cell Biology, Department of Pediatrics, National Jewish Medical and Research Center, Denver, CO
| | - Joanne Domenico
- Division of Cell Biology, Department of Pediatrics, National Jewish Medical and Research Center, Denver, CO
| | - Erwin W. Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Medical and Research Center, Denver, CO
| |
Collapse
|
34
|
|
35
|
Abstract
We propose that retroviruses exploit a cell-encoded pathway of intercellular vesicle traffic, exosome exchange, for both the biogenesis of retroviral particles and a low-efficiency but mechanistically important mode of infection. This Trojan exosome hypothesis reconciles current paradigms of retrovirus-directed transmission with the unique lipid composition of retroviral particles, the host cell proteins present in retroviral particles, the complex cell biology of retroviral release, and the ability of retroviruses to infect cells independently of Envelope protein-receptor interactions. An exosomal origin also predicts that retroviruses pose an unsolvable paradox for adaptive immune responses, that retroviral antigen vaccines are unlikely to provide prophylactic protection, and that alloimmunity is a central component of antiretroviral immunity. Finally, the Trojan exosome hypothesis has important implications for the fight against HIV and AIDS, including how to develop new antiretroviral therapies, assess the risk of retroviral infection, and generate effective antiretroviral vaccines.
Collapse
Affiliation(s)
- Stephen J Gould
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
36
|
Ameisen JC, Lelièvre JD, Pleskoff O. HIV/host interactions: new lessons from the Red Queen's country. AIDS 2003; 16 Suppl 4:S25-31. [PMID: 12698996 DOI: 10.1097/00002030-200216004-00004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jean Claude Ameisen
- EMI-U 9922 INSERM/Université Paris 7, IFR 02, AP-HP, Faculté de Médecine Xavier Bichat, 16 rue Henri Huchard, 75877 Paris cedex 18, France.
| | | | | |
Collapse
|