1
|
Guajardo-Contreras GI, Abdalla AL, Chen A, Niu M, Beauchamp E, Berthiaume LG, Cochrane AW, Mouland AJ. HIV-1 N-myristoylation-dependent hijacking of late endosomes/lysosomes to drive Gag assembly in macrophages. J Cell Sci 2024; 137:jcs263588. [PMID: 39439384 DOI: 10.1242/jcs.263588] [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: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Macrophages represent an important viral reservoir in HIV-1-infected individuals. Different from T cells, HIV-1 assembly in macrophages occurs at intracellular compartments termed virus-containing compartments (VCCs). Our previous research in HeLa cells - in which assembly resembles that found in infected T cells - suggested that late endosomes/lysosomes (LELs) play a role in HIV-1 trafficking towards its assembly sites. However, the role of LELs during assembly at VCCs is not fully understood. Herein, we used the HIV-1-inducible cell line THP-1 GagZip as a model to study HIV-1 Gag intracellular trafficking and assembly in macrophages. We demonstrated LEL involvement at VCCs using various microscopy techniques and biochemical approaches. Live-cell imaging revealed that HIV-1 repositions LELs towards the plasma membrane and modulates their motility. We showed that Arl8b-mediated LEL repositioning is not responsible for Gag trafficking to VCCs. Additionally, the inhibition of myristoylation by PCLX-001 decreased the presence of Gag on endosomes and inhibited VCC formation in both the THP-1 cell line and primary macrophages. In conclusion, we present evidence supporting the idea that HIV-1 manipulates the LEL trajectory to guide Gag to VCCs in an N-myristoylation-dependent manner.
Collapse
Affiliation(s)
- Gabriel I Guajardo-Contreras
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Lady Davis Institute at the Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Ana L Abdalla
- Lady Davis Institute at the Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alex Chen
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Meijuan Niu
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | | | - Luc G Berthiaume
- Pacylex Pharmaceuticals Inc., Edmonton, AB T5J 4P6, Canada
- Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Alan W Cochrane
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Andrew J Mouland
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
2
|
Viral and Host Factors Regulating HIV-1 Envelope Protein Trafficking and Particle Incorporation. Viruses 2022; 14:v14081729. [PMID: 36016351 PMCID: PMC9415270 DOI: 10.3390/v14081729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
The HIV-1 envelope glycoprotein (Env) is an essential structural component of the virus, serving as the receptor-binding protein and principal neutralizing determinant. Env trimers are incorporated into developing particles at the plasma membrane of infected cells. Incorporation of HIV-1 Env into particles in T cells and macrophages is regulated by the long Env cytoplasmic tail (CT) and the matrix region of Gag. The CT incorporates motifs that interact with cellular factors involved in endosomal trafficking. Env follows an unusual pathway to arrive at the site of particle assembly, first traversing the secretory pathway to the plasma membrane (PM), then undergoing endocytosis, followed by directed sorting to the site of particle assembly on the PM. Many aspects of Env trafficking remain to be defined, including the sequential events that occur following endocytosis, leading to productive recycling and particle incorporation. This review focuses on the host factors and pathways involved in Env trafficking, and discusses leading models of Env incorporation into particles.
Collapse
|
3
|
Lerner G, Weaver N, Anokhin B, Spearman P. Advances in HIV-1 Assembly. Viruses 2022; 14:v14030478. [PMID: 35336885 PMCID: PMC8952333 DOI: 10.3390/v14030478] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 12/10/2022] Open
Abstract
The assembly of HIV-1 particles is a concerted and dynamic process that takes place on the plasma membrane of infected cells. An abundance of recent discoveries has advanced our understanding of the complex sequence of events leading to HIV-1 particle assembly, budding, and release. Structural studies have illuminated key features of assembly and maturation, including the dramatic structural transition that occurs between the immature Gag lattice and the formation of the mature viral capsid core. The critical role of inositol hexakisphosphate (IP6) in the assembly of both the immature and mature Gag lattice has been elucidated. The structural basis for selective packaging of genomic RNA into virions has been revealed. This review will provide an overview of the HIV-1 assembly process, with a focus on recent advances in the field, and will point out areas where questions remain that can benefit from future investigation.
Collapse
|
4
|
Zhang W, Bai J, Hang K, Xu J, Zhou C, Li L, Wang Z, Wang Y, Wang K, Xue D. Role of Lysosomal Acidification Dysfunction in Mesenchymal Stem Cell Senescence. Front Cell Dev Biol 2022; 10:817877. [PMID: 35198560 PMCID: PMC8858834 DOI: 10.3389/fcell.2022.817877] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has been widely used as a potential treatment for a variety of diseases. However, the contradiction between the low survival rate of transplanted cells and the beneficial therapeutic effects has affected its clinical use. Lysosomes as organelles at the center of cellular recycling and metabolic signaling, play essential roles in MSC homeostasis. In the first part of this review, we summarize the role of lysosomal acidification dysfunction in MSC senescence. In the second part, we summarize some of the potential strategies targeting lysosomal proteins to enhance the therapeutic effect of MSCs.
Collapse
Affiliation(s)
- Weijun Zhang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinwu Bai
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kai Hang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianxiang Xu
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengwei Zhou
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lijun Li
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongxiang Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yibo Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kanbin Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Deting Xue
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Deting Xue,
| |
Collapse
|
5
|
Thomas JR, Naidu P, Appios A, McGovern N. The Ontogeny and Function of Placental Macrophages. Front Immunol 2021; 12:771054. [PMID: 34745147 PMCID: PMC8566952 DOI: 10.3389/fimmu.2021.771054] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/05/2021] [Indexed: 01/02/2023] Open
Abstract
The placenta is a fetal-derived organ whose function is crucial for both maternal and fetal health. The human placenta contains a population of fetal macrophages termed Hofbauer cells. These macrophages play diverse roles, aiding in placental development, function and defence. The outer layer of the human placenta is formed by syncytiotrophoblast cells, that fuse to form the syncytium. Adhered to the syncytium at sites of damage, on the maternal side of the placenta, is a population of macrophages termed placenta associated maternal macrophages (PAMM1a). Here we discuss recent developments that have led to renewed insight into our understanding of the ontogeny, phenotype and function of placental macrophages. Finally, we discuss how the application of new technologies within placental research are helping us to further understand these cells.
Collapse
Affiliation(s)
| | | | | | - Naomi McGovern
- Department of Pathology and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Basyuk E, Rage F, Bertrand E. RNA transport from transcription to localized translation: a single molecule perspective. RNA Biol 2021; 18:1221-1237. [PMID: 33111627 PMCID: PMC8354613 DOI: 10.1080/15476286.2020.1842631] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/21/2022] Open
Abstract
Transport of mRNAs is an important step of gene expression, which brings the genetic message from the DNA in the nucleus to a precise cytoplasmic location in a regulated fashion. Perturbation of this process can lead to pathologies such as developmental and neurological disorders. In this review, we discuss recent advances in the field of mRNA transport made using single molecule fluorescent imaging approaches. We present an overview of these approaches in fixed and live cells and their input in understanding the key steps of mRNA journey: transport across the nucleoplasm, export through the nuclear pores and delivery to its final cytoplasmic location. This review puts a particular emphasis on the coupling of mRNA transport with translation, such as localization-dependent translational regulation and translation-dependent mRNA localization. We also highlight the recently discovered translation factories, and how cellular and viral RNAs can hijack membrane transport systems to travel in the cytoplasm.
Collapse
Affiliation(s)
- Eugenia Basyuk
- Institut de Génétique Humaine, CNRS-UMR9002, Univ Montpellier, Montpellier, France
- Present address: Laboratoire de Microbiologie Fondamentale et Pathogénicité, CNRS-UMR 5234, Université de Bordeaux, Bordeaux, France
| | - Florence Rage
- Institut de Génétique Moléculaire de Montpellier, CNRS-UMR5535, Univ Montpellier, Montpellier, France
| | - Edouard Bertrand
- Institut de Génétique Humaine, CNRS-UMR9002, Univ Montpellier, Montpellier, France
- Institut de Génétique Moléculaire de Montpellier, CNRS-UMR5535, Univ Montpellier, Montpellier, France
- Equipe Labélisée Ligue Nationale Contre Le Cancer, Montpellier, France
| |
Collapse
|
7
|
Hendricks CM, Cordeiro T, Gomes AP, Stevenson M. The Interplay of HIV-1 and Macrophages in Viral Persistence. Front Microbiol 2021; 12:646447. [PMID: 33897659 PMCID: PMC8058371 DOI: 10.3389/fmicb.2021.646447] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/11/2021] [Indexed: 12/18/2022] Open
Abstract
HIV-1 has evolved mechanisms to evade host cell immune responses and persist for lifelong infection. Latent cellular reservoirs are responsible for this persistence of HIV-1 despite the powerful effects of highly active antiretroviral therapies (HAART) to control circulating viral load. While cellular reservoirs have been extensively studied, much of these studies have focused on peripheral blood and resting memory CD4+ T cells containing latent HIV-1 provirus; however, efforts to eradicate cellular reservoirs have been stunted by reservoirs found in tissues compartments that are not easily accessible. These tissues contain resting memory CD4+ T cells and tissue resident macrophages, another latent cellular reservoir to HIV-1. Tissue resident macrophages have been associated with HIV-1 infection since the 1980s, and evidence has continued to grow regarding their role in HIV-1 persistence. Specific biological characteristics play a vital role as to why macrophages are latent cellular reservoirs for HIV-1, and in vitro and in vivo studies exhibit how macrophages contribute to viral persistence in individuals and animals on antiretroviral therapies. In this review, we characterize the role and evolutionary advantages of macrophage reservoirs to HIV-1 and their contribution to HIV-1 persistence. In acknowledging the interplay of HIV-1 and macrophages in the host, we identify reasons why current strategies are incapable of eliminating HIV-1 reservoirs and why efforts must focus on eradicating reservoirs to find a future functional cure.
Collapse
Affiliation(s)
- Chynna M Hendricks
- Department of Microbiology & Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Thaissa Cordeiro
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ana Paula Gomes
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Mario Stevenson
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
8
|
Veenhuis RT, Abreu CM, Shirk EN, Gama L, Clements JE. HIV replication and latency in monocytes and macrophages. Semin Immunol 2021; 51:101472. [PMID: 33648815 PMCID: PMC10171083 DOI: 10.1016/j.smim.2021.101472] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 02/20/2021] [Indexed: 12/13/2022]
Abstract
The relevance of monocyte and macrophage reservoirs in virally suppressed people with HIV (vsPWH) has previously been debatable. Macrophages were assumed to have a moderate life span and lack self-renewing potential. However, recent studies have challenged this dogma and now suggest an important role of these cell as long-lived HIV reservoirs. Lentiviruses have a long-documented association with macrophages and abundant evidence exists that macrophages are important target cells for HIV in vivo. A critical understanding of HIV infection, replication, and latency in macrophages is needed in order to determine the appropriate method of measuring and eliminating this cellular reservoir. This review provides a brief discussion of the biology and acute and chronic infection of monocytes and macrophages, with a more substantial focus on replication, latency and measurement of the reservoir in cells of myeloid origin.
Collapse
Affiliation(s)
- Rebecca T Veenhuis
- Department of Molecular and Comparative Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Celina M Abreu
- Department of Molecular and Comparative Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Erin N Shirk
- Department of Molecular and Comparative Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lucio Gama
- Department of Molecular and Comparative Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| | - Janice E Clements
- Department of Molecular and Comparative Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
9
|
Clayton KL, Mylvaganam G, Villasmil-Ocando A, Stuart H, Maus MV, Rashidian M, Ploegh HL, Walker BD. HIV-infected macrophages resist efficient NK cell-mediated killing while preserving inflammatory cytokine responses. Cell Host Microbe 2021; 29:435-447.e9. [PMID: 33571449 DOI: 10.1016/j.chom.2021.01.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/19/2020] [Accepted: 01/12/2021] [Indexed: 12/30/2022]
Abstract
Natural killer (NK) cells are innate cytolytic effectors that target HIV-infected CD4+ T cells. In conjunction with antibodies recognizing the HIV envelope, NK cells also eliminate HIV-infected targets through antibody-dependent cellular cytotoxicity (ADCC). However, how these NK cell functions impact infected macrophages is less understood. We show that HIV-infected macrophages resist NK cell-mediated killing. Compared with HIV-infected CD4+ T cells, initial innate NK cell interactions with HIV-infected macrophages skew the response toward cytokine production, rather than release of cytolytic contents, causing inefficient elimination of infected macrophages. Studies with chimeric antigen receptor (CAR) T cells demonstrate that the viral envelope is equally accessible on CD4+ T cells and macrophages. Nonetheless, ADCC against macrophages is muted compared with ADCC against CD4+ T cells. Thus, HIV-infected macrophages employ mechanisms to evade immediate cytolytic NK cell function while preserving inflammatory cytokine responses. These findings emphasize the importance of eliminating infected macrophages for HIV cure efforts.
Collapse
Affiliation(s)
- Kiera L Clayton
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Geetha Mylvaganam
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | | | - Heather Stuart
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | - Mohammad Rashidian
- Dana-Farber Cancer Institute, Boston, MA 02215, USA; Boston Children's Hospital, Boston, MA 02115, USA
| | - Hidde L Ploegh
- Boston Children's Hospital, Boston, MA 02115, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Massachusetts General Hospital, Boston, MA 02114, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Institute of Medical Engineering and Sciences and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02138, USA.
| |
Collapse
|
10
|
Rai MA, Hammonds J, Pujato M, Mayhew C, Roskin K, Spearman P. Comparative analysis of human microglial models for studies of HIV replication and pathogenesis. Retrovirology 2020; 17:35. [PMID: 33213476 PMCID: PMC7678224 DOI: 10.1186/s12977-020-00544-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Background HIV associated neurocognitive disorders cause significant morbidity and mortality despite the advent of highly active antiretroviral therapy. A deeper understanding of fundamental mechanisms underlying HIV infection and pathogenesis in the central nervous system is warranted. Microglia are resident myeloid cells of the brain that are readily infected by HIV and may constitute a CNS reservoir. We evaluated two microglial model cell lines (C20, HMC3) and two sources of primary cell-derived microglia (monocyte-derived microglia [MMG] and induced pluripotent stem cell-derived microglia [iPSC-MG]) as potential model systems for studying HIV-microglia interactions. Results All four microglial model cells expressed typical myeloid markers with the exception of low or absent CD45 and CD11b expression by C20 and HMC3, and all four expressed the microglia-specific markers P2RY12 and TMEM119. Marked differences were observed upon gene expression profiling, however, indicating that MMG and iPSC-MG cluster closely together with primary human microglial cells, while C20 and HMC3 were similar to each other but very different from primary microglia. Expression of HIV-relevant genes also revealed important differences, with iPSC-MG and MMG expressing relevant genes at levels more closely resembling primary microglia. iPSC-MG and MMG were readily infected with R5-tropic HIV, while C20 and HMC3 lack CD4 and require pseudotyping for infection. Despite many similarities, HIV replication dynamics and HIV-1 particle capture by Siglec-1 differed markedly between the MMG and iPSC-MG. Conclusions MMG and iPSC-MG appear to be viable microglial models that are susceptible to HIV infection and bear more similarities to authentic microglia than two transformed microglia cell lines. The observed differences in HIV replication and particle capture between MMG and iPSC-MG warrant further study.
Collapse
Affiliation(s)
- Mohammad A Rai
- Division of Infectious Diseases, Cincinnati Children's Hospital, 3333 Burnet Avenue, MLC 7017, Cincinnati, OH, 45229, USA.,Division of Infectious Diseases, Department of Medicine, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jason Hammonds
- Division of Infectious Diseases, Cincinnati Children's Hospital, 3333 Burnet Avenue, MLC 7017, Cincinnati, OH, 45229, USA.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Mario Pujato
- Division of Biomedical Informatics, Cincinnati Children's Hospital, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Christopher Mayhew
- Pluripotent Stem Cell Core Facility, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Krishna Roskin
- Division of Biomedical Informatics, Cincinnati Children's Hospital, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Paul Spearman
- Division of Infectious Diseases, Cincinnati Children's Hospital, 3333 Burnet Avenue, MLC 7017, Cincinnati, OH, 45229, USA. .,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
11
|
Ins and Outs of Reovirus: Vesicular Trafficking in Viral Entry and Egress. Trends Microbiol 2020; 29:363-375. [PMID: 33008713 PMCID: PMC7523517 DOI: 10.1016/j.tim.2020.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
Cell entry and egress are essential steps in the viral life cycle that govern pathogenesis and spread. Mammalian orthoreoviruses (reoviruses) are nonenveloped viruses implicated in human disease that serve as tractable models for studies of pathogen-host interactions. In this review we discuss the function of intracellular vesicular transport systems in reovirus entry, trafficking, and egress and comment on shared themes for diverse viruses. Designing strategic therapeutic interventions that impede these steps in viral replication requires a detailed understanding of mechanisms by which viruses coopt vesicular trafficking. We illuminate such targets, which may foster development of antiviral agents.
Collapse
|
12
|
Auld SC, Staitieh BS. HIV and the tuberculosis "set point": how HIV impairs alveolar macrophage responses to tuberculosis and sets the stage for progressive disease. Retrovirology 2020; 17:32. [PMID: 32967690 PMCID: PMC7509826 DOI: 10.1186/s12977-020-00540-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/15/2020] [Indexed: 11/16/2022] Open
Abstract
As HIV has fueled a global resurgence of tuberculosis over the last several decades, there is a growing awareness that HIV-mediated impairments in both innate and adaptive immunity contribute to the heightened risk of tuberculosis in people with HIV. Since early immune responses to Mycobacterium tuberculosis (Mtb) set the stage for subsequent control or progression to active tuberculosis disease, early host-pathogen interactions following Mtb infection can be thought of as establishing a mycobacterial "set point," which we define as the mycobacterial burden at the point of adaptive immune activation. This early immune response is impaired in the context of HIV coinfection, allowing for a higher mycobacterial set point and greater likelihood of progression to active disease with greater bacterial burden. Alveolar macrophages, as the first cells to encounter Mtb in the lungs, play a critical role in containing Mtb growth and establishing the mycobacterial set point. However, a number of key macrophage functions, ranging from pathogen recognition and uptake to phagocytosis and microbial killing, are blunted in HIV coinfection. To date, research evaluating the effects of HIV on the alveolar macrophage response to Mtb has been relatively limited, particularly with regard to the critical early events that help to dictate the mycobacterial set point. A greater understanding of alveolar macrophage functions impacted by HIV coinfection will improve our understanding of protective immunity to Mtb and may reveal novel pathways amenable to intervention to improve both early immune control of Mtb and clinical outcomes for the millions of people worldwide infected with HIV.
Collapse
Affiliation(s)
- Sara C Auld
- Emory University School of Medicine, Atlanta, GA, USA.
- Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | | |
Collapse
|
13
|
Gea-Mallorquí E, Zablocki-Thomas L, Maurin M, Jouve M, Rodrigues V, Ruffin N, Benaroch P. HIV-2-Infected Macrophages Produce and Accumulate Poorly Infectious Viral Particles. Front Microbiol 2020; 11:1603. [PMID: 32754142 PMCID: PMC7365954 DOI: 10.3389/fmicb.2020.01603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/18/2020] [Indexed: 12/30/2022] Open
Abstract
A significant proportion of HIV-2-infected patients exhibit natural virological control that is generally absent from HIV-1-infected patients. Along with CD4+ T cells, HIV-1 targets macrophages which may contribute to viral spreading and the latent reservoir. We have studied the relationship between macrophages and HIV-2, focusing on post-entry steps. HIV-2-infected monocyte-derived macrophages (MDMs) produced substantial amounts of viral particles that were largely harbored intracellularly. New viruses assembled at the limiting membrane of internal compartments similar to virus-containing compartments (VCCs) described for HIV-1. VCCs from MDMs infected with either virus shared protein composition and morphology. Strikingly, HIV-2 Gag was mostly absent from the cytosol and almost exclusively localized to the VCCs, whereas HIV-1 Gag was distributed in both locations. Ultrastructural analyses of HIV-2-infected MDMs revealed the presence of numerous VCCs containing both immature and mature particles in the lumen. HIV-2 particles produced de novo by MDMs were poorly infectious in reporter cells and in transmission to activated T cells through a process that appeared independent of BST2 restriction. Rather than being involved in viral spreading, HIV-2-infected macrophages may represent a cell-associated source of viral antigens that can participate in the immune control of HIV-2 infection.
Collapse
Affiliation(s)
| | | | - Mathieu Maurin
- Institut Curie, PSL∗ Research University, INSERM U932, Paris, France
| | - Mabel Jouve
- Institut Curie, PSL∗ Research University, UMR3216, Paris, France
| | - Vasco Rodrigues
- Institut Curie, PSL∗ Research University, INSERM U932, Paris, France
| | - Nicolas Ruffin
- Institut Curie, PSL∗ Research University, INSERM U932, Paris, France
| | - Philippe Benaroch
- Institut Curie, PSL∗ Research University, INSERM U932, Paris, France
| |
Collapse
|
14
|
Dupont M, Sattentau QJ. Macrophage Cell-Cell Interactions Promoting HIV-1 Infection. Viruses 2020; 12:E492. [PMID: 32354203 PMCID: PMC7290394 DOI: 10.3390/v12050492] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Many pathogens infect macrophages as part of their intracellular life cycle. This is particularly true for viruses, of which HIV-1 is one of the best studied. HIV-1 infection of macrophages has important consequences for viral persistence and pathogenesis, but the mechanisms of macrophage infection remain to be fully elucidated. Despite expressing viral entry receptors, macrophages are inefficiently infected by cell-free HIV-1 virions, whereas direct cell-cell spread is more efficient. Different modes of cell-cell spread have been described, including the uptake by macrophages of infected T cells and the fusion of infected T cells with macrophages, both leading to macrophage infection. Cell-cell spread can also transmit HIV-1 between macrophages and from macrophages to T cells. Here, we describe the current state of the field concerning the cell-cell spread of HIV-1 to and from macrophages, discuss mechanisms, and highlight potential in vivo relevance.
Collapse
Affiliation(s)
- Maeva Dupont
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford OX13RE, UK
| | | |
Collapse
|
15
|
Strategies employed by viruses to manipulate autophagy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:203-237. [PMID: 32620243 DOI: 10.1016/bs.pmbts.2020.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy, originally described as a conserved bulk degradation pathway important to maintain cellular homeostasis during starvation, has also been implicated in playing a central role in multiple physiological processes. For example, autophagy is part of our innate immunity by targeting intracellular pathogens to lysosomes for degradation in a process called xenophagy. Coevolution and adaptation between viruses and autophagy have armed viruses with a multitude of strategies to counteract the antiviral functions of the autophagy pathway. In addition, some viruses have acquired mechanisms to exploit specific functions of either autophagy or the key components of this process, the autophagy-related (ATG) proteins, to promote viral replication and pathogenesis. In this chapter, we describe several examples where the strategy employed by a virus to subvert autophagy has been described with molecular detail. Their stratagems positively or negatively target practically all the steps of autophagy, including the signaling pathways regulating this process. This highlights the intricate relationship between autophagy and viruses and how by commandeering autophagy, viruses have devised ways to fine-tune their replication.
Collapse
|
16
|
Lepont L, Leymarie O, Berlioz-Torrent C. [Cellular and viral determinants control HIV-1 virus-containing compartment expansion in infected macrophages]. Med Sci (Paris) 2020; 36:20-23. [PMID: 32014092 DOI: 10.1051/medsci/2019255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Leslie Lepont
- Université de Paris, Institut Cochin, Inserm, CNRS, laboratoire « Interactions hôte-virus », 27 rue du faubourg Saint Jacques, F-75014 Paris, France
| | - Olivier Leymarie
- Université de Paris, Institut Cochin, Inserm, CNRS, laboratoire « Interactions hôte-virus », 27 rue du faubourg Saint Jacques, F-75014 Paris, France
| | - Clarisse Berlioz-Torrent
- Université de Paris, Institut Cochin, Inserm, CNRS, laboratoire « Interactions hôte-virus », 27 rue du faubourg Saint Jacques, F-75014 Paris, France
| |
Collapse
|
17
|
Constitutive Siglec-1 expression confers susceptibility to HIV-1 infection of human dendritic cell precursors. Proc Natl Acad Sci U S A 2019; 116:21685-21693. [PMID: 31591213 DOI: 10.1073/pnas.1911007116] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The human dendritic cell (DC) lineage has recently been unraveled by high-dimensional mapping, revealing the existence of a discrete new population of blood circulating DC precursors (pre-DCs). Whether this new DC population possesses specific functional features as compared to the other blood DC subset upon pathogen encounter remained to be evaluated. A unique feature of pre-DCs among blood DCs is their constitutive expression of the viral adhesion receptor Siglec-1. Here, we show that pre-DCs, but not other blood DC subsets, are susceptible to infection by HIV-1 in a Siglec-1-dependent manner. Siglec-1 mediates pre-DC infection of CCR5- and CXCR4-tropic strains. Infection of pre-DCs is further enhanced in the presence of HIV-2/SIVmac Vpx, indicating that Siglec-1 does not counteract restriction factors such as SAMHD1. Instead, Siglec-1 promotes attachment and fusion of viral particles. HIV-1-infected pre-DCs produce new infectious viral particles that accumulate in intracellular compartments reminiscent of the virus-containing compartment of macrophages. Pre-DC activation by toll-like receptor (TLR) ligands induces an antiviral state that inhibits HIV-1 fusion and infection, but Siglec-1 remains functional and mediates replication-independent transfer of HIV-1 to activated primary T lymphocytes. Altogether, Siglec-1-mediated susceptibility to HIV-1 infection of pre-DCs constitutes a unique functional feature that might represent a preferential relationship of this emerging cell type with viruses.
Collapse
|
18
|
Contribution of the Cytoplasmic Determinants of Vpu to the Expansion of Virus-Containing Compartments in HIV-1-Infected Macrophages. J Virol 2019; 93:JVI.00020-19. [PMID: 30867316 DOI: 10.1128/jvi.00020-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/09/2019] [Indexed: 12/30/2022] Open
Abstract
HIV-1 infection of macrophages leads to the sequestration of newly formed viruses in intracellular plasma membrane-connected structures termed virus-containing compartments (VCCs), where virions remain infectious and hidden from immune surveillance. The cellular restriction factor bone marrow stromal cell antigen 2 (BST2), which prevents HIV-1 dissemination by tethering budding viral particles at the plasma membrane, can be found in VCCs. The HIV-1 accessory protein Vpu counteracts the restriction factor BST2 by downregulating its expression and removing it from viral budding sites. Numerous studies described these Vpu countermeasures in CD4+ T cells or model cell lines, but the interplay between Vpu and BST2 in VCC formation and HIV-1 production in macrophages is less explored. Here, we show that Vpu expression in HIV-1-infected macrophages enhances viral release. This effect is related to Vpu's ability to circumvent BST2 antiviral activity. We show that in absence of Vpu, BST2 is enriched in VCCs and colocalizes with capsid p24, whereas Vpu expression significantly reduces the presence of BST2 in these compartments. Furthermore, our data reveal that BST2 is dispensable for the formation of VCCs and that Vpu expression impacts the volume of these compartments. This Vpu activity partly depends on BST2 expression and requires the integrity of the Vpu transmembrane domain, the dileucine-like motif E59XXXLV64 and phosphoserines 52 and 56 of Vpu. Altogether, these results highlight that Vpu controls the volume of VCCs and promotes HIV-1 release from infected macrophages.IMPORTANCE HIV-1 infection of macrophages leads to the sequestration of newly formed viruses in virus-containing compartments (VCCs), where virions remain infectious and hidden from immune surveillance. The restriction factor BST2, which prevents HIV-1 dissemination by tethering budding viral particles, can be found in VCCs. The HIV-1 Vpu protein counteracts BST2. This study explores the interplay between Vpu and BST2 in the viral protein functions on HIV-1 release and viral particle sequestration in VCCs in macrophages. The results show that Vpu controls the volume of VCCs and favors viral particle release. These Vpu functions partly depend on Vpu's ability to antagonize BST2. This study highlights that the transmembrane domain of Vpu and two motifs of the Vpu cytoplasmic domain are required for these functions. These motifs were notably involved in the control of the volume of VCCs by Vpu but were dispensable for the prevention of the specific accumulation of BST2 in these structures.
Collapse
|
19
|
Cellular Determinants of HIV Persistence on Antiretroviral Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1075:213-239. [PMID: 30030795 DOI: 10.1007/978-981-13-0484-2_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The era of antiretroviral therapy has made HIV-1 infection a manageable chronic disease for those with access to treatment. Despite treatment, virus persists in tissue reservoirs seeded with long-lived infected cells that are resistant to cell death and immune recognition. Which cells contribute to this reservoir and which factors determine their persistence are central questions that need to be answered to achieve viral eradication. In this chapter, we describe how cell susceptibility to infection, resistance to cell death, and immune-mediated killing as well as natural cell life span and turnover potential are central components that allow persistence of different lymphoid and myeloid cell subsets that were recently identified as key players in harboring latent and actively replicating virus. The relative contribution of these subsets to persistence of viral reservoir is described, and the open questions are highlighted.
Collapse
|
20
|
Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 2019; 21:9-17. [PMID: 30602770 DOI: 10.1038/s41556-018-0250-9] [Citation(s) in RCA: 2431] [Impact Index Per Article: 405.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023]
Abstract
The ability of exosomes to transfer cargo from donor to acceptor cells, thereby triggering phenotypic changes in the latter, has generated substantial interest in the scientific community. However, the extent to which exosomes differ from other extracellular vesicles in terms of their biogenesis and functions remains ill-defined. Here, we discuss the current knowledge on the specificities of exosomes and other types of extracellular vesicles, and their roles as important agents of cell-to-cell communication.
Collapse
|
21
|
Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 2019. [PMID: 30602770 DOI: 10.1038/s41556-018-0250-259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2023]
Abstract
The ability of exosomes to transfer cargo from donor to acceptor cells, thereby triggering phenotypic changes in the latter, has generated substantial interest in the scientific community. However, the extent to which exosomes differ from other extracellular vesicles in terms of their biogenesis and functions remains ill-defined. Here, we discuss the current knowledge on the specificities of exosomes and other types of extracellular vesicles, and their roles as important agents of cell-to-cell communication.
Collapse
Affiliation(s)
- Mathilde Mathieu
- Institut Curie, PSL Research University, INSERM U932, Paris, France
- Université Paris Descartes, Paris, France
| | | | - Grégory Lavieu
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932, Paris, France.
| |
Collapse
|
22
|
Zhang Y, Lu J, Ma J, Liu X. Insulin-induced gene 1 (INSIG1) inhibits HIV-1 production by degrading Gag via activity of the ubiquitin ligase TRC8. J Biol Chem 2018; 294:2046-2059. [PMID: 30563842 DOI: 10.1074/jbc.ra118.004630] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/04/2018] [Indexed: 11/06/2022] Open
Abstract
Insulin-induced gene 1 (INSIG1) regulates sterol synthesis by mediating the activation of sterol regulatory element-binding protein (SREBP) and the degradation of the HMG-CoA reductase (HMGCR). INSIG1 is up-regulated during HIV-1 infection, but its role in HIV-1 infection is unknown. In this report, using pseudovirus production, protein overexpression, and gene knockouts, we found that INSIG1 inhibits HIV-1 production by accelerating the degradation of the HIV-1 Gag protein. Unlike the degradation of HMGCR via the E3 ubiquitin ligase autocrine motility factor receptor (AMFR), a process that depends on the proteasome, INSIG1 coordinated with another ligase, translocation in renal carcinoma chromosome 8 (TRC8), and promoted Gag degradation through the lysosome pathway. We conclude that INSIG1 functions as a sentinel responsive to HIV-1 production and inhibits HIV-1 replication by degrading Gag, a process occurring at intracellular membrane sites such as the endoplasmic reticulum and endosomes where both INSIG1 and Gag may be located.
Collapse
Affiliation(s)
- You Zhang
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jing Lu
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jing Ma
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinqi Liu
- From the State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
23
|
Dimethyl Fumarate Prevents HIV-Induced Lysosomal Dysfunction and Cathepsin B Release from Macrophages. J Neuroimmune Pharmacol 2018; 13:345-354. [PMID: 29987592 DOI: 10.1007/s11481-018-9794-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 05/28/2018] [Indexed: 12/17/2022]
Abstract
HIV-associated neurocognitive disorders (HAND) are prevalent despite combined antiretroviral therapy, affecting nearly half of HIV-infected patients worldwide. During HIV infection of macrophages secretion of the lysosomal protein, cathepsin B, is increased. Secreted cathepsin B has been shown to induce neurotoxicity. Oxidative stress is increased in HIV-infected patients, while antioxidants are decreased in monocytes from patients with HIV-associated dementia (HAD). Dimethyl fumarate (DMF), an antioxidant, has been reported to decrease HIV replication and neurotoxicity mediated by HIV-infected macrophages. Thus, we hypothesized that DMF will decrease cathepsin B release from HIV-infected macrophages by preventing oxidative stress and enhancing lysosomal function. Monocyte-derived macrophages (MDM) were isolated from healthy donors, inoculated with HIV-1ADA, and treated with DMF following virus removal. After 12 days post-infection, HIV-1 p24 and total cathepsin B levels were measured from HIV-infected MDM supernatants using ELISA; intracellular reactive oxygen and nitrogen species (ROS/RNS) were measured from MDM lysates, and functional lysosomes were assessed using a pH-dependent lysosomal dye. Neurons were incubated with serum-free conditioned media from DMF-treated MDM and neurotoxicity was determined using TUNEL assay. Results indicate that DMF reduced HIV-1 replication and cathepsin B secretion from HIV-infected macrophages in a dose-dependent manner. Also, DMF decreased intracellular ROS/RNS levels, and prevented HIV-induced lysosomal dysfunction and neuronal apoptosis. In conclusion, the improvement in lysosomal function with DMF treatment may represent the possible mechanism to reduce HIV-1 replication and cathepsin B secretion. DMF represents a potential therapeutic strategy against HAND.
Collapse
|
24
|
García M, Buzón MJ, Benito JM, Rallón N. Peering into the HIV reservoir. Rev Med Virol 2018; 28:e1981. [PMID: 29744964 DOI: 10.1002/rmv.1981] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022]
Abstract
The main obstacle to HIV eradication is the establishment of a long-term persistent HIV reservoir. Although several therapeutic approaches have been developed to reduce and eventually eliminate the HIV reservoir, only a few have achieved promising results. A better knowledge of the mechanisms involved in the establishment and maintenance of HIV reservoir is of utmost relevance for the design of new therapeutic strategies aimed at purging it with the ultimate goal of achieving HIV eradication or alternatively a functional cure. In this regard, it is also important to take a close look into the cellular HIV reservoirs other than resting memory CD4 T-cells with key roles in reservoir maintenance that have been recently described. Unraveling the special characteristics of these HIV cellular compartments could aid us in designing new therapeutic strategies to deplete the latent HIV reservoir.
Collapse
Affiliation(s)
- Marcial García
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | | | - José M Benito
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - Norma Rallón
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| |
Collapse
|
25
|
Slow Release of HIV-1 Protein Nef from Vesicle-like Structures Is Inhibited by Cytosolic Calcium Elevation in Single Human Microglia. Mol Neurobiol 2018; 56:102-118. [DOI: 10.1007/s12035-018-1072-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/09/2018] [Indexed: 12/14/2022]
|
26
|
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
|
27
|
T Cell-Macrophage Fusion Triggers Multinucleated Giant Cell Formation for HIV-1 Spreading. J Virol 2017; 91:JVI.01237-17. [PMID: 28978713 DOI: 10.1128/jvi.01237-17] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/29/2017] [Indexed: 01/05/2023] Open
Abstract
HIV-1-infected macrophages participate in virus dissemination and establishment of virus reservoirs in host tissues, but the mechanisms for virus cell-to-cell transfer to macrophages remain unknown. Here, we reveal the mechanisms for cell-to-cell transfer from infected T cells to macrophages and virus spreading between macrophages. We show that contacts between infected T lymphocytes and macrophages lead to cell fusion for the fast and massive transfer of CCR5-tropic viruses to macrophages. Through the merge of viral material between T cells and macrophages, these newly formed lymphocyte-macrophage fused cells acquire the ability to fuse with neighboring noninfected macrophages. Together, these two-step envelope-dependent cell fusion processes lead to the formation of highly virus-productive multinucleated giant cells reminiscent of the infected multinucleated giant macrophages detected in HIV-1-infected patients and simian immunodeficiency virus-infected macaques. These mechanisms represent an original mode of virus transmission for viral spreading and a new model for the formation of macrophage virus reservoirs during infection.IMPORTANCE We reveal a very efficient mechanism involved in cell-to-cell transfer from infected T cells to macrophages and subsequent virus spreading between macrophages by a two-step cell fusion process. Infected T cells first establish contacts and fuse with macrophage targets. The newly formed lymphocyte-macrophage fused cells then acquire the ability to fuse with surrounding uninfected macrophages, leading to the formation of infected multinucleated giant cells that can survive for a long time, as evidenced in vivo in lymphoid organs and the central nervous system. This route of infection may be a major determinant for virus dissemination and the formation of macrophage virus reservoirs in host tissues during HIV-1 infection.
Collapse
|
28
|
Rodrigues V, Ruffin N, San-Roman M, Benaroch P. Myeloid Cell Interaction with HIV: A Complex Relationship. Front Immunol 2017; 8:1698. [PMID: 29250073 PMCID: PMC5714857 DOI: 10.3389/fimmu.2017.01698] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022] Open
Abstract
Cells of the myeloid lineage, particularly macrophages, serve as primary hosts for HIV in vivo, along with CD4 T lymphocytes. Macrophages are present in virtually every tissue of the organism, including locations with negligible T cell colonization, such as the brain, where HIV-mediated inflammation may lead to pathological sequelae. Moreover, infected macrophages are present in multiple other tissues. Recent evidence obtained in humanized mice and macaque models highlighted the capacity of macrophages to sustain HIV replication in vivo in the absence of T cells. Combined with the known resistance of the macrophage to the cytopathic effects of HIV infection, such data bring a renewed interest in this cell type both as a vehicle for viral spread as well as a viral reservoir. While our understanding of key processes of HIV infection of macrophages is far from complete, recent years have nevertheless brought important insight into the uniqueness of the macrophage infection. Productive infection of macrophages by HIV can occur by different routes including from phagocytosis of infected T cells. In macrophages, HIV assembles and buds into a peculiar plasma membrane-connected compartment that preexists to the infection. While the function of such compartment remains elusive, it supposedly allows for the persistence of infectious viral particles over extended periods of time and may play a role on viral transmission. As cells of the innate immune system, macrophages have the capacity to detect and respond to viral components. Recent data suggest that such sensing may occur at multiple steps of the viral cycle and impact subsequent viral spread. We aim to provide an overview of the HIV-macrophage interaction along the multiple stages of the viral life cycle, extending when pertinent such observations to additional myeloid cell types such as dendritic cells or blood monocytes.
Collapse
Affiliation(s)
- Vasco Rodrigues
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Nicolas Ruffin
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Mabel San-Roman
- Institut Curie, PSL Research University, UMR3216, Paris, France
| | | |
Collapse
|
29
|
Yaseen MM, Abuharfeil NM, Yaseen MM, Shabsoug BM. The role of polymorphonuclear neutrophils during HIV-1 infection. Arch Virol 2017; 163:1-21. [PMID: 28980078 DOI: 10.1007/s00705-017-3569-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/14/2017] [Indexed: 12/23/2022]
Abstract
It is well-recognized that human immunodeficiency virus type-1 (HIV-1) mainly targets CD4+ T cells and macrophages. Nonetheless, during the past three decades, a huge number of studies have reported that HIV-1 can directly or indirectly target other cellular components of the immune system including CD8+ T cells, B cells, dendritic cells, natural killer cells, and polymorphonuclear neutrophils (PMNs), among others. PMNs are the most abundant leukocytes in the human circulation, and are known to play principal roles in the elimination of invading pathogens, regulating different immune responses, healing of injured tissues, and maintaining mucosal homeostasis. Until recently, little was known about the impact of HIV-1 infection on PMNs as well as the impact of PMNs on HIV-1 disease progression. This is because early studies focused on neutropenia and recurrent microbial infections, particularly, during advanced disease. However, recent studies have extended the investigation area to cover new aspects of the interactions between HIV-1 and PMNs. This review aims to summarize these advances and address the impact of HIV-1 infection on PMNs as well as the impact of PMNs on HIV-1 disease progression to better understand the pathophysiology of HIV-1 infection.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Medical Laboratory Sciences, College of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Applied Biological Sciences, College of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad Mahmoud Yaseen
- Public Health, College of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Barakat Mohammad Shabsoug
- Chemical Sciences, College of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
30
|
Durantel D, Kusters I, Louis J, Manel N, Ottenhoff THM, Picot V, Saaadatian-Elahi M. Mechanisms behind TB, HBV, and HIV chronic infections. INFECTION GENETICS AND EVOLUTION 2017; 55:142-150. [PMID: 28919545 DOI: 10.1016/j.meegid.2017.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/13/2022]
Abstract
Immune evasion is critical for pathogens to maintain their presence within hosts, giving rise to chronic infections. Here, we examine the immune evasion strategies employed by three pathogens with high medical burden, namely, tuberculosis, HIV and HBV. Establishment of chronic infection by these pathogens is a multi-step process that involves an interplay between restriction factor, innate immunity and adaptive immunity. Engagement of these host defences is intimately linked with specific steps within the pathogen replication cycles. Critical host factors are increasingly recognized to regulate immune evasion and susceptibility to disease. Fuelled by innovative technology development, the understanding of these mechanisms provides critical knowledge for rational design of vaccines and therapeutic immune strategies.
Collapse
Affiliation(s)
- David Durantel
- Cancer Research Center of Lyon (CRCL), INSERM, U1052, CNRS, University of Lyon, UMR_5286, LabEx DEVweCAN, Lyon, France
| | - Inca Kusters
- Sanofi Pasteur, 2 Avenue du Pont Pasteur, 69367 Lyon Cedex 07, France
| | - Jacques Louis
- Fondation Mérieux, 17 rue Bourgelat, 69002 Lyon, France
| | - Nicolas Manel
- Immunity and Cancer Department, Institute Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Tom H M Ottenhoff
- Group Immunology and Immunogenetics of Bacterial Infectious Diseases, Dept. of Infectious Diseases, Leiden University Medical Center, Bldg. 1, Rm # C-05-43 Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | | | - Mitra Saaadatian-Elahi
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, 5 Place d'Arsonval, 69437 Lyon Cedex 03, France.
| |
Collapse
|
31
|
Kubo Y, Masumoto H, Izumida M, Kakoki K, Hayashi H, Matsuyama T. Rab3a-Bound CD63 Is Degraded and Rab3a-Free CD63 Is Incorporated into HIV-1 Particles. Front Microbiol 2017; 8:1653. [PMID: 28900422 PMCID: PMC5581869 DOI: 10.3389/fmicb.2017.01653] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/15/2017] [Indexed: 01/23/2023] Open
Abstract
CD63, a member of the tetraspanin family, is involved in virion production by human immunodeficiency virus type 1 (HIV-1), but its mechanism is unknown. In this study, we showed that a small GTP-binding protein, Rab3a, interacts with CD63. When Rab3a was exogenously expressed, the amounts of CD63 decreased in cells. The Rab3a-mediated reduction of CD63 was suppressed by lysosomal and proteasomal inhibitors. The amount of CD63 was increased by reducing the endogenous Rab3a level using a specific shRNA. These results indicate that Rab3a binds to CD63 to induce the degradation of CD63. Rab3a is thought to be involved in exocytosis, but we found that another function of Rab3a affects the fate of CD63 in lysosomes. CD63 interacted with Rab3a and was incorporated into HIV-1 particles. However, Rab3a was not detected in HIV-1 virions, thereby indicating that Rab3a-free CD63, but not Rab3a-bound CD63, is incorporated into HIV-1 particles. Overexpression or silencing of Rab3a moderately reduced HIV-1 virion formation. Overexpression of Rab3a decreased CD63 levels, but did not affect the incorporation of CD63 into HIV-1 particles. This study showed that Rab3a binds to CD63 to induce the degradation of CD63, and only Rab3a-free CD63 is incorporated into HIV-1 particles.
Collapse
Affiliation(s)
- Yoshinao Kubo
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasaki, Japan.,Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasaki, Japan
| | - Hiroshi Masumoto
- Biomedical Research Support Center, Nagasaki University School of MedicineNagasaki, Japan
| | - Mai Izumida
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasaki, Japan.,Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki UniversityNagasaki, Japan
| | - Katsura Kakoki
- Department of Urology, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasaki, Japan
| | - Hideki Hayashi
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasaki, Japan.,Medical University Research Administrator, Nagasaki University School of MedicineNagasaki, Japan
| | - Toshifumi Matsuyama
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasaki, Japan
| |
Collapse
|
32
|
The Role of Caveolin 1 in HIV Infection and Pathogenesis. Viruses 2017; 9:v9060129. [PMID: 28587148 PMCID: PMC5490806 DOI: 10.3390/v9060129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/02/2017] [Accepted: 05/22/2017] [Indexed: 12/29/2022] Open
Abstract
Caveolin 1 (Cav-1) is a major component of the caveolae structure and is expressed in a variety of cell types including macrophages, which are susceptible to human immunodeficiency virus (HIV) infection. Caveolae structures are present in abundance in mechanically stressed cells such as endothelial cells and adipocytes. HIV infection induces dysfunction of these cells and promotes pathogenesis. Cav-1 and the caveolae structure are believed to be involved in multiple cellular processes that include signal transduction, lipid regulation, endocytosis, transcytosis, and mechanoprotection. Such a broad biological role of Cav-1/caveolae is bound to have functional cross relationships with several molecular pathways including HIV replication and viral-induced pathogenesis. The current review covers the relationship of Cav-1 and HIV in respect to viral replication, persistence, and the potential role in pathogenesis.
Collapse
|
33
|
Rizkallah G, Alais S, Futsch N, Tanaka Y, Journo C, Mahieux R, Dutartre H. Dendritic cell maturation, but not type I interferon exposure, restricts infection by HTLV-1, and viral transmission to T-cells. PLoS Pathog 2017; 13:e1006353. [PMID: 28426803 PMCID: PMC5413061 DOI: 10.1371/journal.ppat.1006353] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/02/2017] [Accepted: 04/13/2017] [Indexed: 12/21/2022] Open
Abstract
Human T lymphotropic Virus type 1 (HTLV-1) is the etiological agent of Adult T cell Leukemia/Lymphoma (ATLL) and HTLV-1-Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). Both CD4+ T-cells and dendritic cells (DCs) infected with HTLV-1 are found in peripheral blood from HTLV-1 carriers. We previously demonstrated that monocyte-derived IL-4 DCs are more susceptible to HTLV-1 infection than autologous primary T-cells, suggesting that DC infection precedes T-cell infection. However, during blood transmission, breast-feeding or sexual transmission, HTLV-1 may encounter different DC subsets present in the blood, the intestinal or genital mucosa respectively. These different contacts may impact HTLV-1 ability to infect DCs and its subsequent transfer to T-cells. Using in vitro monocyte-derived IL-4 DCs, TGF-β DCs and IFN-α DCs that mimic DCs contacting HTLV-1 in vivo, we show here that despite their increased ability to capture HTLV-1 virions, IFN-α DCs restrict HTLV-1 productive infection. Surprisingly, we then demonstrate that it is not due to the antiviral activity of type-I interferon produced by IFN-α DCs, but that it is likely to be linked to a distinct trafficking route of HTLV-1 in IL-4 DCs vs. IFN-α DCs. Finally, we demonstrate that, in contrast to IL-4 DCs, IFN-α DCs are impaired in their capacity to transfer HTLV-1 to CD4 T-cells, both after viral capture and trans-infection and after their productive infection. In conclusion, the nature of the DCs encountered by HTLV-1 upon primo-infection and the viral trafficking route through the vesicular pathway of these cells determine the efficiency of viral transmission to T-cells, which may condition the fate of infection.
Collapse
Affiliation(s)
- Gergès Rizkallah
- International Center for Research in Infectiology, Retroviral Oncogenesis laboratory, INSERM U1111 –Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Equipe labellisée “Ligue Nationale Contre le Cancer”, Lyon, France
| | - Sandrine Alais
- International Center for Research in Infectiology, Retroviral Oncogenesis laboratory, INSERM U1111 –Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Equipe labellisée “Ligue Nationale Contre le Cancer”, Lyon, France
| | - Nicolas Futsch
- International Center for Research in Infectiology, Retroviral Oncogenesis laboratory, INSERM U1111 –Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Equipe labellisée “Ligue Nationale Contre le Cancer”, Lyon, France
| | - Yuetsu Tanaka
- Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Uehara 207, Nishihara-cho, Okinawa, Japan
| | - Chloé Journo
- International Center for Research in Infectiology, Retroviral Oncogenesis laboratory, INSERM U1111 –Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Equipe labellisée “Ligue Nationale Contre le Cancer”, Lyon, France
| | - Renaud Mahieux
- International Center for Research in Infectiology, Retroviral Oncogenesis laboratory, INSERM U1111 –Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Equipe labellisée “Ligue Nationale Contre le Cancer”, Lyon, France
| | - Hélène Dutartre
- International Center for Research in Infectiology, Retroviral Oncogenesis laboratory, INSERM U1111 –Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Equipe labellisée “Ligue Nationale Contre le Cancer”, Lyon, France
| |
Collapse
|
34
|
Hammonds JE, Beeman N, Ding L, Takushi S, Francis AC, Wang JJ, Melikyan GB, Spearman P. Siglec-1 initiates formation of the virus-containing compartment and enhances macrophage-to-T cell transmission of HIV-1. PLoS Pathog 2017; 13:e1006181. [PMID: 28129379 PMCID: PMC5298340 DOI: 10.1371/journal.ppat.1006181] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 02/08/2017] [Accepted: 01/12/2017] [Indexed: 11/22/2022] Open
Abstract
HIV-1 particles assemble and bud from the plasma membrane of infected T lymphocytes. Infected macrophages, in contrast, accumulate particles within an apparent intracellular compartment known as the virus-containing compartment or VCC. Many aspects of the formation and function of the VCC remain unclear. Here we demonstrate that VCC formation does not actually require infection of the macrophage, but can be reproduced through the exogenous addition of non-infectious virus-like particles or infectious virions to macrophage cultures. Particles were captured by Siglec-1, a prominent cell surface lectin that attaches to gangliosides on the lipid envelope of the virus. VCCs formed within infected macrophages were readily targeted by the addition of ganglioside-containing virus-like particles to the extracellular media. Depletion of Siglec-1 from the macrophage or depletion of gangliosides from viral particles prevented particle uptake into the VCC and resulted in substantial reductions of VCC volume. Furthermore, Siglec-1-mediated virion capture and subsequent VCC formation was required for efficient trans-infection of autologous T cells. Our results help to define the nature of this intracellular compartment, arguing that it is a compartment formed by particle uptake from the periphery, and that this compartment can readily transmit virus to target T lymphocytes. Inhibiting or eliminating the VCC may be an important component of strategies to reduce HIV transmission and to eradicate HIV reservoirs. T lymphocytes and macrophages are the two major cell types involved in HIV replication and transmission events. When a T cell is infected, virus particles assemble and bud from the plasma membrane of the cell. In contrast, infected macrophages develop an intracellular collection of viruses termed the virus-containing compartment or VCC. Many aspects of the formation and function of the VCC remain unclear. Here we show that VCC formation does not actually require infection of the macrophage, but can be reproduced through the addition of virus-like particles or infectious virions to macrophages. HIV-1 particles were captured by the cell surface carbohydrate-binding protein Siglec-1, followed by co-migration of Siglec-1 and captured viral particles to the VCC. Depletion of Siglec-1 from the macrophage prevented VCC formation, and inhibited the ability of infected macrophages to transmit HIV to T cells. Our results help to define the origin of this intracellular compartment, arguing that it is a compartment formed by particle uptake from the periphery. Inhibiting or eliminating the VCC may be an important component of strategies to reduce HIV transmission and to eradicate HIV reservoirs.
Collapse
Affiliation(s)
- Jason E. Hammonds
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Neal Beeman
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Lingmei Ding
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Sarah Takushi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ashwanth C. Francis
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jaang-Jiun Wang
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Gregory B. Melikyan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Paul Spearman
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
35
|
Inlora J, Chukkapalli V, Bedi S, Ono A. Molecular Determinants Directing HIV-1 Gag Assembly to Virus-Containing Compartments in Primary Macrophages. J Virol 2016; 90:8509-19. [PMID: 27440886 PMCID: PMC5021390 DOI: 10.1128/jvi.01004-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/11/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED The subcellular sites of HIV-1 assembly, determined by the localization of the structural protein Gag, vary in a cell-type-dependent manner. In T cells and transformed cell lines used as model systems, HIV-1 assembles at the plasma membrane (PM). The binding and localization of HIV-1 Gag to the PM are mediated by the interaction between the matrix (MA) domain, specifically the highly basic region, and a PM-specific acidic phospholipid, phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2]. In primary macrophages, prominent accumulation of assembling or assembled particles is found in the virus-containing compartments (VCCs), which largely consist of convoluted invaginations of the PM. To elucidate the molecular mechanism of HIV-1 Gag targeting to the VCCs, we examined the impact of overexpression of polyphosphoinositide 5-phosphatase IV (5ptaseIV), which depletes cellular PI(4,5)P2, in primary macrophages. We found that the VCC localization and virus release of HIV-1 are severely impaired upon 5ptaseIV overexpression, suggesting an important role for the MA-PI(4,5)P2 interaction in HIV-1 assembly in primary macrophages. However, our analysis of HIV-1 Gag derivatives with MA changes showed that this interaction contributes to Gag membrane binding but is dispensable for specific targeting of Gag to the VCCs per se We further determined that deletion of the NC domain abolishes VCC-specific localization of HIV-1 Gag. Notably, HIV-1 Gag localized efficiently to the VCCs when the NC domain was replaced with a leucine zipper dimerization motif that promotes Gag multimerization. Altogether, our data revealed that targeting of HIV-1 Gag to the VCCs requires NC-dependent multimerization. IMPORTANCE In T cells and model cell lines, HIV-1 Gag localizes to the PM in a manner dependent on the MA-PI(4,5)P2 interaction. On the other hand, in primary macrophages, HIV-1 Gag localizes to convoluted intracellular membrane structures termed virus-containing compartments (VCCs). Although these compartments have been known for decades, and despite the implication of viruses in VCCs being involved in virus reservoir maintenance and spread, the viral determinant(s) that promotes Gag targeting to VCCs is unknown. In this study, we found that the MA-PI(4,5)P2 interaction facilitates efficient Gag membrane binding in macrophages but is not essential for Gag targeting to VCCs. Rather, our results revealed that NC-dependent multimerization promotes VCC targeting. Our findings highlight the differential roles played by MA and NC in HIV-1 Gag membrane binding and targeting and suggest a multimerization-dependent mechanism for Gag trafficking in primary macrophages similar to that for Gag localization to uropods in polarized T cells.
Collapse
Affiliation(s)
- Jingga Inlora
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Vineela Chukkapalli
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sukhmani Bedi
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
36
|
The Envelope Cytoplasmic Tail of HIV-1 Subtype C Contributes to Poor Replication Capacity through Low Viral Infectivity and Cell-to-Cell Transmission. PLoS One 2016; 11:e0161596. [PMID: 27598717 PMCID: PMC5012655 DOI: 10.1371/journal.pone.0161596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/08/2016] [Indexed: 12/15/2022] Open
Abstract
The cytoplasmic tail (gp41CT) of the HIV-1 envelope (Env) mediates Env incorporation into virions and regulates Env intracellular trafficking. Little is known about the functional impact of variability in this domain. To address this issue, we compared the replication of recombinant virus pairs carrying the full Env (Env viruses) or the Env ectodomain fused to the gp41CT of NL4.3 (EnvEC viruses) (12 subtype C and 10 subtype B pairs) in primary CD4+ T-cells and monocyte-derived-macrophages (MDMs). In CD4+ T-cells, replication was as follows: B-EnvEC = B-Env>C-EnvEC>C-Env, indicating that the gp41CT of subtype C contributes to the low replicative capacity of this subtype. In MDMs, in contrast, replication capacity was comparable for all viruses regardless of subtype and of gp41CT. In CD4+ T-cells, viral entry, viral release and viral gene expression were similar. However, infectivity of free virions and cell-to-cell transmission of C-Env viruses released by CD4+ T-cells was lower, suggestive of lower Env incorporation into virions. Subtype C matrix only minimally rescued viral replication and failed to restore infectivity of free viruses and cell-to-cell transmission. Taken together, these results show that polymorphisms in the gp41CT contribute to viral replication capacity and suggest that the number of Env spikes per virion may vary across subtypes. These findings should be taken into consideration in the design of vaccines.
Collapse
|
37
|
Cell-Free versus Cell-to-Cell Infection by Human Immunodeficiency Virus Type 1 and Human T-Lymphotropic Virus Type 1: Exploring the Link among Viral Source, Viral Trafficking, and Viral Replication. J Virol 2016; 90:7607-17. [PMID: 27334587 DOI: 10.1128/jvi.00407-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) and human T-lymphotropic virus type 1 (HTLV-1) are complex retroviruses mainly infecting CD4(+) T lymphocytes. In addition, antigen-presenting cells such as dendritic cells (DCs) are targeted in vivo by both viruses, although to a lesser extent. Interaction of HIV-1 with DCs plays a key role in viral dissemination from the mucosa to CD4(+) T lymphocytes present in lymphoid organs. While similar mechanisms may occur for HTLV-1 as well, most HTLV-1 data were obtained from T-cell studies, and little is known regarding the trafficking of this virus in DCs. We first compared the efficiency of cell-free versus cell-associated viral sources of both retroviruses at infecting DCs. We showed that both HIV-1 and HTLV-1 cell-free particles are poorly efficient at productively infecting DCs, except when DC-SIGN has been engaged. Furthermore, while SAMHD-1 accounts for restriction of cell-free HIV-1 infection, it is not involved in HTLV-1 restriction. In addition, cell-free viruses lead mainly to a nonproductive DC infection, leading to trans-infection of T-cells, a process important for HIV-1 spread but not for that of HTLV-1. Finally, we show that T-DC cell-to-cell transfer implies viral trafficking in vesicles that may both increase productive infection of DCs ("cis-infection") and allow viral escape from immune surveillance. Altogether, these observations allowed us to draw a model of HTLV-1 and HIV-1 trafficking in DCs.
Collapse
|
38
|
Human Immunodeficiency Virus Type 2 (HIV-2) Gag Is Trafficked in an AP-3 and AP-5 Dependent Manner. PLoS One 2016; 11:e0158941. [PMID: 27392064 PMCID: PMC4938559 DOI: 10.1371/journal.pone.0158941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/24/2016] [Indexed: 02/03/2023] Open
Abstract
Although human immunodeficiency virus (HIV) types 1 and 2 are closely related lentiviruses with similar replication cycles, HIV-2 infection is associated with slower progression to AIDS, a higher proportion of long term non-progressors, and lower rates of transmission than HIV-1, likely as a consequence of a lower viral load during HIV-2 infection. A mechanistic explanation for the differential viral load remains unclear but knowledge of differences in particle production between HIV-1 and HIV-2 may help to shed light on this issue. In contrast to HIV-1, little is known about the assembly of HIV-2 particles, and the trafficking of HIV-2 Gag, the structural component of the virus, within cells. We have established that HIV-2 Gag accumulates in intracellular CD63 positive compartments, from which it may be delivered or recycled to the cell surface, or degraded. HIV-2 particle release was dependent on the adaptor protein complex AP-3 and the newly identified AP-5 complex, but much less so on AP-1. In contrast, HIV-1 particle release required AP-1 and AP-3, but not AP-5. AP-2, an essential component of clathrin-mediated endocytosis, which was previously shown to be inhibitory to HIV-1 particle release, had no effect on HIV-2. The differential requirement for adaptor protein complexes confirmed that HIV-1 and HIV-2 Gag have distinct cellular trafficking pathways, and that HIV-2 particles may be more susceptible to degradation prior to release.
Collapse
|
39
|
Nkwe DO, Pelchen-Matthews A, Burden JJ, Collinson LM, Marsh M. The intracellular plasma membrane-connected compartment in the assembly of HIV-1 in human macrophages. BMC Biol 2016; 14:50. [PMID: 27338237 PMCID: PMC4919869 DOI: 10.1186/s12915-016-0272-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/09/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In HIV-infected macrophages, newly formed progeny virus particles accumulate in intracellular plasma membrane-connected compartments (IPMCs). Although the virus is usually seen in these compartments, it is unclear whether HIV assembly is specifically targeted to IPMCs or whether some viruses may also form at the cell surface but are not detected, as particles budding from the latter site will be released into the medium. RESULTS To investigate the fidelity of HIV-1 targeting to IPMCs compared to the cell surface directly, we generated mutants defective in recruitment of the Endosomal Sorting Complexes Required for Transport (ESCRT) proteins required for virus scission. For mutants unable to bind the ESCRT-I component Tsg101, HIV release was inhibited and light and electron microscopy revealed that budding was arrested. When expressed in human monocyte-derived macrophages (MDM), these mutants formed budding-arrested, immature particles at their assembly sites, allowing us to capture virtually all of the virus budding events. A detailed morphological analysis of the distribution of the arrested viruses by immunofluorescence staining and confocal microscopy, and by electron microscopy, demonstrated that HIV assembly in MDMs is targeted primarily to IPMCs, with fewer than 5 % of budding events seen at the cell surface. Morphometric analysis of the relative membrane areas at the cell surface and IPMCs confirmed a large enrichment of virus assembly events in IPMCs. Serial block-face scanning electron microscopy of macrophages infected with a budding-defective HIV mutant revealed high-resolution 3D views of the complex organisation of IPMCs, with in excess of 15,000 associated HIV budding sites, and multiple connections between IPMCs and the cell surface. CONCLUSIONS Using detailed quantitative analysis, we demonstrate that HIV assembly in MDMs is specifically targeted to IPMCs. Furthermore, 3D analysis shows, for the first time, the detailed ultrastructure of an IPMC within a large cell volume, at a resolution that allowed identification of individual virus assembly events, and potential portals through which virus may be released during cell-cell transfer. These studies provide new insights to the organisation of the HIV assembly compartments in macrophages, and show how HIV particles accumulating in these protected sites may function as a virus reservoir.
Collapse
Affiliation(s)
- David O. Nkwe
- />MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT UK
- />Present Address: Department of Biology and Biotechnological Sciences, College of Science, Botswana International University of Science and Technology, Private Bag 16, Palapye, Botswana
| | - Annegret Pelchen-Matthews
- />MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Jemima J. Burden
- />MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Lucy M. Collinson
- />The Francis Crick Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London, WC2A 3LY UK
| | - Mark Marsh
- />MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| |
Collapse
|
40
|
Abstract
Lentiviruses have a long-documented association with macrophages. Abundant evidence exists for in vitro and, in a tissue-specific manner, in vivo infection of macrophages by the primate lentiviruses HIV-1 and SIV. However, macrophage contribution to aspects of HIV-1 and SIV pathogenesis, and their role in viral persistence in individuals on suppressive antiretroviral therapy, remains unclear. Here we discuss recent evidence implicating macrophages in HIV-1-mediated disease and highlight directions for further investigation.
Collapse
|
41
|
Dumas A, Lê-Bury G, Marie-Anaïs F, Herit F, Mazzolini J, Guilbert T, Bourdoncle P, Russell DG, Benichou S, Zahraoui A, Niedergang F. The HIV-1 protein Vpr impairs phagosome maturation by controlling microtubule-dependent trafficking. J Cell Biol 2016; 211:359-72. [PMID: 26504171 PMCID: PMC4621833 DOI: 10.1083/jcb.201503124] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The HIV protein Vpr interacts with EB1, p150Glued, and dynein heavy chain and perturbs the centripetal movement of phagosomes and their maturation, resulting in impaired phagolysosome biogenesis, which is important for bacterial clearance and cytokine production. Human immunodeficiency virus type 1 (HIV-1) impairs major functions of macrophages but the molecular basis for this defect remains poorly characterized. Here, we show that macrophages infected with HIV-1 were unable to respond efficiently to phagocytic triggers and to clear bacteria. The maturation of phagosomes, defined by the presence of late endocytic markers, hydrolases, and reactive oxygen species, was perturbed in HIV-1–infected macrophages. We showed that maturation arrest occurred at the level of the EHD3/MICAL-L1 endosomal sorting machinery. Unexpectedly, we found that the regulatory viral protein (Vpr) was crucial to perturb phagosome maturation. Our data reveal that Vpr interacted with EB1, p150Glued, and dynein heavy chain and was sufficient to critically alter the microtubule plus end localization of EB1 and p150Glued, hence altering the centripetal movement of phagosomes and their maturation. Thus, we identify Vpr as a modulator of the microtubule-dependent endocytic trafficking in HIV-1–infected macrophages, leading to strong alterations in phagolysosome biogenesis.
Collapse
Affiliation(s)
- Audrey Dumas
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Gabrielle Lê-Bury
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Florence Marie-Anaïs
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Floriane Herit
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Julie Mazzolini
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Thomas Guilbert
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Pierre Bourdoncle
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Serge Benichou
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Ahmed Zahraoui
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Florence Niedergang
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France Centre National de la Recherche Scientifique UMR 8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| |
Collapse
|
42
|
HIV-1 strategies to overcome the immune system by evading and invading innate immune system. HIV & AIDS REVIEW 2016. [DOI: 10.1016/j.hivar.2015.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
43
|
Human Immunodeficiency Virus Type 1 Nef Inhibits Autophagy through Transcription Factor EB Sequestration. PLoS Pathog 2015; 11:e1005018. [PMID: 26115100 PMCID: PMC4482621 DOI: 10.1371/journal.ppat.1005018] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/09/2015] [Indexed: 11/19/2022] Open
Abstract
HIV Nef acts as an anti-autophagic maturation factor through interaction with beclin-1 (BECN1). We report that exposure of macrophages to infectious or non-infectious purified HIV induces toll-like receptor 8 (TLR8) and BECN1 dependent dephosphorylation and nuclear translocation of TFEB and that this correlates with an increase in autophagy markers. RNA interference for ATG13, TFEB, TLR8, or BECN1 inhibits this HIV-induced autophagy. However, once HIV establishes a productive infection, TFEB phosphorylation and cytoplasmic sequestration are increased resulting in decreased autophagy markers. Moreover, by 7 d post-infection, autophagy levels are similar to mock infected controls. Conversely, although Nef deleted HIV similarly induces TFEB dephosphorylation and nuclear localization, and increases autophagy, these levels remain elevated during continued productive infection. Thus, the interaction between HIV and TLR8 serves as a signal for autophagy induction that is dependent upon the dephosphorylation and nuclear translocation of TFEB. During permissive infection, Nef binds BECN1 resulting in mammalian target of rapamycin (MTOR) activation, TFEB phosphorylation and cytosolic sequestration, and the inhibition of autophagy. To our knowledge, this is the first report of a virus modulating TFEB localization and helps to explain how HIV modulates autophagy to promote its own replication and cell survival. Under basal conditions, the mammalian target of rapamycin (MTOR) phosphorylates transcription factor EB (TFEB) resulting in its cytoplasmic retention. When MTOR is inhibited, TFEB is dephosphorylated and translocated to the nucleus where it increases autophagy and lysosomal gene expression. As human immunodeficiency virus type 1 (HIV) Nef acts as an anti-autophagic maturation factor through interaction with beclin-1 (BECN1), we investigated the role of Nef and TFEB in the modulation of autophagy during HIV infection of human macrophages. We found that upon exposure to HIV, macrophages elicited an autophagic response through a toll-like receptor 8 (TLR8) and BECN1 dependent dephosphorylation and nuclear translocation of TFEB. However, once HIV infection is established, phosphorylation and cytoplasmic sequestration of TFEB as well as autophagy revert to pre-infection levels. Moreover, this reversion is dependent upon the presence of HIV Nef. Collectively, the data suggests that the interaction between HIV and TLR8 serves as a signal for autophagy induction that is dependent upon the dephosphorylation and nuclear translocation of TFEB. Once HIV establishes a productive infection, Nef binds BECN1 resulting in MTOR activation, TFEB phosphorylation and cytosolic sequestration and the inhibition of autophagy.
Collapse
|
44
|
Involvement of the Rac1-IRSp53-Wave2-Arp2/3 Signaling Pathway in HIV-1 Gag Particle Release in CD4 T Cells. J Virol 2015; 89:8162-81. [PMID: 26018170 DOI: 10.1128/jvi.00469-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 05/14/2015] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED During HIV-1 assembly, the Gag viral proteins are targeted and assemble at the inner leaflet of the cell plasma membrane. This process could modulate the cortical actin cytoskeleton, located underneath the plasma membrane, since actin dynamics are able to promote localized membrane reorganization. In addition, activated small Rho GTPases are known for regulating actin dynamics and membrane remodeling. Therefore, the modulation of such Rho GTPase activity and of F-actin by the Gag protein during virus particle formation was considered. Here, we studied the implication of the main Rac1, Cdc42, and RhoA small GTPases, and some of their effectors, in this process. The effect of small interfering RNA (siRNA)-mediated Rho GTPases and silencing of their effectors on Gag localization, Gag membrane attachment, and virus-like particle production was analyzed by immunofluorescence coupled to confocal microscopy, membrane flotation assays, and immunoblot assays, respectively. In parallel, the effect of Gag expression on the Rac1 activation level was monitored by G-LISA, and the intracellular F-actin content in T cells was monitored by flow cytometry and fluorescence microscopy. Our results revealed the involvement of activated Rac1 and of the IRSp53-Wave2-Arp2/3 signaling pathway in HIV-1 Gag membrane localization and particle release in T cells as well as a role for actin branching and polymerization, and this was solely dependent on the Gag viral protein. In conclusion, our results highlight a new role for the Rac1-IRSp53-Wave2-Arp2/3 signaling pathway in the late steps of HIV-1 replication in CD4 T lymphocytes. IMPORTANCE During HIV-1 assembly, the Gag proteins are targeted and assembled at the inner leaflet of the host cell plasma membrane. Gag interacts with specific membrane phospholipids that can also modulate the regulation of cortical actin cytoskeleton dynamics. Actin dynamics can promote localized membrane reorganization and thus can be involved in facilitating Gag assembly and particle formation. Activated small Rho GTPases and effectors are regulators of actin dynamics and membrane remodeling. We thus studied the effects of the Rac1, Cdc42, and RhoA GTPases and their specific effectors on HIV-1 Gag membrane localization and viral particle release in T cells. Our results show that activated Rac1 and the IRSp53-Wave2-Arp2/3 signaling pathway are involved in Gag plasma membrane localization and viral particle production. This work uncovers a role for cortical actin through the activation of Rac1 and the IRSp53/Wave2 signaling pathway in HIV-1 particle formation in CD4 T lymphocytes.
Collapse
|
45
|
Gerber PP, Cabrini M, Jancic C, Paoletti L, Banchio C, von Bilderling C, Sigaut L, Pietrasanta LI, Duette G, Freed EO, Basile GDS, Moita CF, Moita LF, Amigorena S, Benaroch P, Geffner J, Ostrowski M. Rab27a controls HIV-1 assembly by regulating plasma membrane levels of phosphatidylinositol 4,5-bisphosphate. J Cell Biol 2015; 209:435-52. [PMID: 25940347 PMCID: PMC4427790 DOI: 10.1083/jcb.201409082] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 04/01/2015] [Indexed: 12/22/2022] Open
Abstract
During the late stages of the HIV-1 replication cycle, the viral polyprotein Pr55(Gag) is recruited to the plasma membrane (PM), where it binds phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and directs HIV-1 assembly. We show that Rab27a controls the trafficking of late endosomes carrying phosphatidylinositol 4-kinase type 2 α (PI4KIIα) toward the PM of CD4(+) T cells. Hence, Rab27a promotes high levels of PM phosphatidylinositol 4-phosphate and the localized production of PI(4,5)P2, therefore controlling Pr55(Gag) membrane association. Rab27a also controls PI(4,5)P2 levels at the virus-containing compartments of macrophages. By screening Rab27a effectors, we identified that Slp2a, Slp3, and Slac2b are required for the association of Pr55(Gag) with the PM and that Slp2a cooperates with Rab27a in the recruitment of PI4KIIα to the PM. We conclude that by directing the trafficking of PI4KIIα-positive endosomes toward the PM, Rab27a controls PI(4,5)P2 production and, consequently, HIV-1 replication.
Collapse
Affiliation(s)
- Pehuén Pereyra Gerber
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Mercedes Cabrini
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Carolina Jancic
- Instituto de Medicina Experimental-CONICET, Academia Nacional de Medicina, C1425AUM Buenos Aires, Argentina
| | - Luciana Paoletti
- Instituto de Biologia Molecular y Celular de Rosario-CONICET, S2000EZP Santa Fe, Argentina
| | - Claudia Banchio
- Instituto de Biologia Molecular y Celular de Rosario-CONICET, S2000EZP Santa Fe, Argentina
| | - Catalina von Bilderling
- Instituto de Física de Buenos Aires-CONICET, Departamento de Física; and Centro de Microscopías Avanzadas; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
| | - Lorena Sigaut
- Instituto de Física de Buenos Aires-CONICET, Departamento de Física; and Centro de Microscopías Avanzadas; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
| | - Lía I Pietrasanta
- Instituto de Física de Buenos Aires-CONICET, Departamento de Física; and Centro de Microscopías Avanzadas; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
| | - Gabriel Duette
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Eric O Freed
- Virus-Cell Interaction Section, HIV Drug Resistance Program, National Cancer Institute, Frederick, MD 21702
| | - Genevieve de Saint Basile
- Institut National de la Santé et de la Recherche Médicale U768 and Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, 75015 Paris, France Institut National de la Santé et de la Recherche Médicale U768 and Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, 75015 Paris, France
| | - Catarina Ferreira Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciencia, 2780-156 Oeiras, Portugal
| | - Luis Ferreira Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciencia, 2780-156 Oeiras, Portugal
| | - Sebastian Amigorena
- Centre de Recherche, Institut National de la Santé et de la Recherche Médicale U932, Institut Curie, 75248 Paris, France
| | - Philippe Benaroch
- Centre de Recherche, Institut National de la Santé et de la Recherche Médicale U932, Institut Curie, 75248 Paris, France
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Matías Ostrowski
- Instituto de Investigaciones Biomédicas en Retrovirus y Síndrome de Inmunodeficiencia Adquirida (INBIRS)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
46
|
Araínga M, Guo D, Wiederin J, Ciborowski P, McMillan J, Gendelman HE. Opposing regulation of endolysosomal pathways by long-acting nanoformulated antiretroviral therapy and HIV-1 in human macrophages. Retrovirology 2015; 12:5. [PMID: 25608975 PMCID: PMC4307176 DOI: 10.1186/s12977-014-0133-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/19/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Long-acting nanoformulated antiretroviral therapy (nanoART) is designed to improve patient regimen adherence, reduce systemic drug toxicities, and facilitate clearance of human immunodeficiency virus type one (HIV-1) infection. While nanoART establishes drug depots within recycling and late monocyte-macrophage endosomes, whether or not this provides a strategic advantage towards viral elimination has not been elucidated. RESULTS We applied quantitative SWATH-MS proteomics and cell profiling to nanoparticle atazanavir (nanoATV)-treated and HIV-1 infected human monocyte-derived macrophages (MDM). Native ATV and uninfected cells served as controls. Both HIV-1 and nanoATV engaged endolysosomal trafficking for assembly and depot formation, respectively. Notably, the pathways were deregulated in opposing manners by the virus and the nanoATV, likely by viral clearance. Paired-sample z-scores, of the proteomic data sets, showed up- and down- regulation of Rab-linked endolysosomal proteins. NanoART and native ATV treated uninfected cells showed limited effects. The data was confirmed by Western blot. DAVID and KEGG bioinformatics analyses of proteomic data showed relationships between secretory, mobility and phagocytic cell functions and virus and particle trafficking. CONCLUSIONS We posit that modulation of endolysosomal pathways by antiretroviral nanoparticles provides a strategic path to combat HIV infection.
Collapse
Affiliation(s)
- Mariluz Araínga
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Dongwei Guo
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
- />Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Jayme Wiederin
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Pawel Ciborowski
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - JoEllyn McMillan
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E Gendelman
- />Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, Omaha, NE 68198-5880 USA
- />Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
47
|
Placental Hofbauer cells assemble and sequester HIV-1 in tetraspanin-positive compartments that are accessible to broadly neutralizing antibodies. J Int AIDS Soc 2015; 18:19385. [PMID: 25623930 PMCID: PMC4308659 DOI: 10.7448/ias.18.1.19385] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/11/2014] [Accepted: 12/16/2014] [Indexed: 12/23/2022] Open
Abstract
Introduction Within monocyte-derived macrophages, HIV-1 accumulates in intracellular virus-containing compartments (VCCs) that are inaccessible to the external environment, which implicate these cells as latently infected HIV-1 reservoirs. During mother-to-child transmission of HIV-1, human placental macrophages (Hofbauer cells (HCs)) are viral targets, and have been shown to be infected in vivo and sustain low levels of viral replication in vitro; however, the risk of in utero transmission is less than 7%. The role of these primary macrophages as viral reservoirs is largely undefined. The objective of this study is to define potential sites of viral assembly, accumulation and neutralization in HCs given the pivotal role of the placenta in preventing HIV-1 infection in the mother-infant dyad. Methods Term placentae from 20 HIV-1 seronegative women were obtained following caesarian section. VCCs were evaluated by 3D confocal and electron microscopy. Colocalization R values (Pearson's correlation) were quantified with colocalization module of Volocity 5.2.1. Replication kinetics and neutralization studies were evaluated using p24 ELISA. Results We demonstrate that primary HCs assemble and sequester HIV-1BaL in intracellular VCCs, which are enriched in endosomal/lysosomal markers, including CD9, CD81, CD63 and LAMP-1. Following infection, we observed HIV-1 accumulation in potentially acidic compartments, which stained intensely with Lysotracker-Red. Remarkably, these compartments are readily accessible via the cell surface and can be targeted by exogenously applied small molecules and HIV-1-specific broadly neutralizing antibodies. In addition, broadly neutralizing antibodies (4E10 and VRC01) limited viral replication by HIV-1-infected HCs, which may be mediated by FcγRI. Conclusions These findings suggest that placental HCs possess intrinsic adaptations facilitating unique sequestration of HIV-1, and may serve as a protective viral reservoir to permit viral neutralization and/or antiretroviral drug entry in utero.
Collapse
|
48
|
Schachter J, Delgado KV, Barreto-de-Souza V, Bou-Habib DC, Persechini PM, Meyer-Fernandes JR. Inhibition of ecto-ATPase activities impairs HIV-1 infection of macrophages. Immunobiology 2014; 220:589-96. [PMID: 25577295 DOI: 10.1016/j.imbio.2014.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/29/2014] [Accepted: 12/10/2014] [Indexed: 12/21/2022]
Abstract
Nucleotides and nucleosides are secreted into extracellular media at different concentrations as a consequence of different physiologic and pathological conditions. Ecto-nucleotidases, enzymes present on the surface of most cells, hydrolyze these extracellular nucleotides and reduce the concentration of them, thus affecting the activation of different nucleotide and nucleoside receptors. Also, ecto-nucleotidases are present in a number of microorganisms and play important roles in host-pathogen interactions. Here, we characterized the ecto-ATPase activities present on the surface of HIV-1 particle and human macrophages as well. We found that the kinetic properties of HIV-1 and macrophage ecto-ATPases are similar, suggesting that the enzyme is the same. This ecto-ATPase activity was increased in macrophages infected in vitro with HIV-1. Using three different non-related ecto-ATPase inhibitors-POM-1, ARL67156 and BG0-we showed that the inhibition of these macrophage and viral ecto-ATPase activities impairs HIV-1 infection. In addition, we also found that elevated extracellular concentrations of ATP inhibit HIV-1 production by infected macrophages.
Collapse
Affiliation(s)
- Julieta Schachter
- Campus de Xerem, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia para Pesquisa Translacional em Saúde e Ambiente na Região Amazônica (INPeTAm), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Rio de Janeiro, RJ, Brazil.
| | - Kelly Valcárcel Delgado
- Instituto Nacional de Ciência e Tecnologia para Pesquisa Translacional em Saúde e Ambiente na Região Amazônica (INPeTAm), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Rio de Janeiro, RJ, Brazil; Instituto de Biofísica, Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Victor Barreto-de-Souza
- Laboratório de Pesquisa sobre o Timo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratório de Pesquisa sobre o Timo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Pedro Muanis Persechini
- Instituto Nacional de Ciência e Tecnologia para Pesquisa Translacional em Saúde e Ambiente na Região Amazônica (INPeTAm), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Rio de Janeiro, RJ, Brazil; Instituto de Biofísica, Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
49
|
Compton AA, Bruel T, Porrot F, Mallet A, Sachse M, Euvrard M, Liang C, Casartelli N, Schwartz O. IFITM proteins incorporated into HIV-1 virions impair viral fusion and spread. Cell Host Microbe 2014; 16:736-47. [PMID: 25464829 PMCID: PMC7104936 DOI: 10.1016/j.chom.2014.11.001] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/16/2014] [Accepted: 10/15/2014] [Indexed: 10/28/2022]
Abstract
The interferon-induced transmembrane (IFITM) proteins protect cells from diverse virus infections by inhibiting virus-cell fusion. IFITM proteins also inhibit HIV-1 replication through mechanisms only partially understood. We show that when expressed in uninfected lymphocytes, IFITM proteins exert protective effects during cell-free virus infection, but this restriction can be overcome upon HIV-1 cell-to-cell spread. However, when present in virus-producing lymphocytes, IFITM proteins colocalize with viral Env and Gag proteins and incorporate into nascent HIV-1 virions to limit entry into new target cells. IFITM in viral membranes is associated with impaired virion fusion, offering additional and more potent defense against virus spread. Thus, IFITM proteins act additively in both productively infected cells and uninfected target cells to inhibit HIV-1 spread, potentially conferring these proteins with greater breadth and potency against enveloped viruses.
Collapse
Affiliation(s)
- Alex A Compton
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France.
| | - Timothée Bruel
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France
| | - Françoise Porrot
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France
| | - Adeline Mallet
- Institut Pasteur, Imagopole, Ultrastructural Microscopy Platform, Paris 75015, France
| | - Martin Sachse
- Institut Pasteur, Imagopole, Ultrastructural Microscopy Platform, Paris 75015, France
| | - Marine Euvrard
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; École Normale Supérieure, Department of Biology, Cachan 94230, France
| | - Chen Liang
- Lady Davis Institute, McGill AIDS Centre, Montreal, QC H3T 1E2, Canada
| | - Nicoletta Casartelli
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France
| | - Olivier Schwartz
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France; Vaccine Research Institute, Creteil 94010, France.
| |
Collapse
|
50
|
Olety B, Ono A. Roles played by acidic lipids in HIV-1 Gag membrane binding. Virus Res 2014; 193:108-15. [PMID: 24998886 PMCID: PMC4252750 DOI: 10.1016/j.virusres.2014.06.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 06/18/2014] [Accepted: 06/23/2014] [Indexed: 10/25/2022]
Abstract
The MA domain mediates plasma membrane (PM) targeting of HIV-1 Gag, leading to particle assembly at the PM. The interaction between MA and acidic phospholipids, in addition to N-terminal myristoyl moiety, promotes Gag binding to lipid membranes. Among acidic phospholipids, PI(4,5)P2, a PM-specific phosphoinositide, is essential for proper HIV-1 Gag localization to the PM and efficient virus particle production. Recent studies further revealed that MA-bound RNA negatively regulates HIV-1 Gag membrane binding and that PI(4,5)P2 is necessary to overcome this RNA-imposed block. In this review, we will summarize the current understanding of Gag-membrane interactions and discuss potential roles played by acidic phospholipids.
Collapse
Affiliation(s)
- Balaji Olety
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|