1
|
Heeregrave EJ, Thomas J, van Capel TM, de Jong EC, Pollakis G, Paxton WA. Glycan dependent phenotype differences of HIV-1 generated from macrophage versus CD4 + T helper cell populations. Front Immunol 2023; 14:1107349. [PMID: 37415979 PMCID: PMC10320205 DOI: 10.3389/fimmu.2023.1107349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 06/06/2023] [Indexed: 07/08/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) is able to infect a variety of cell types with differences in entry efficiency and replication kinetics determined by the host cell type or the viral phenotype. The phenotype of the virus produced from these various cell types, including infectivity, co-receptor usage and neutralisation sensitivity, may also be affected by the characteristics of the producing cell. This can be due to incorporation of variant cell-specific molecules or differences in post-translational modifications of the gp41/120 envelope. In this study we produced genetically identical virus strains from macrophages, CD4-enriched lymphocytes as well as Th1 and Th2 CD4+ cell lines and compared each different virus stock for their infectivity in various cell types and sensitivity to neutralisation. In order to study the effect of the producer host cell on the virus phenotype, virus stocks were normalised on infectivity and were sequenced to confirm env gene homogeneity. Virus production by Th1 or Th2 cells did not compromise infectivity of the variant cell types tested. We observed no difference in sensitivity to co-receptor blocking agents upon viral passage through Th1 and Th2 CD4+ cell lineages nor did this affect DC-SIGN-mediated viral capture as measured in a transfer assay to CD4+ lymphocytes. Virus produced by macrophages was comparably sensitive to CC-chemokine inhibition as was virus generated from the array of CD4+ lymphocytes. We identified that virus produced from macrophages was fourteen times more resistant to 2G12 neutralisation than virus produced from CD4+ lymphocytes. Macrophage-produced dual-tropic (R5/X4) virus was six times more efficiently transmitted to CD4+ cells than lymphocyte-derived HIV-1 (p<0.0001) after DCSIGN capture. These results provide further insights to what extent the host cell influences viral phenotype and thereby various aspects of HIV-1 pathogenesis but suggest that viruses generated from Th1 versus Th2 cells are consistent in phenotype.
Collapse
Affiliation(s)
- Edwin J. Heeregrave
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology, Institute of Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Toni M. van Capel
- Department of Experimental Immunology, University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Esther C. de Jong
- Department of Experimental Immunology, University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Georgios Pollakis
- Department of Experimental Immunology, University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - William A. Paxton
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical Infection, Microbiology and Immunology, Institute of Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
2
|
T-cell evasion and invasion during HIV-1 infection: The role of HIV-1 Tat protein. Cell Immunol 2022; 377:104554. [DOI: 10.1016/j.cellimm.2022.104554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022]
|
3
|
Ma T, Luo X, George AF, Mukherjee G, Sen N, Spitzer TL, Giudice LC, Greene WC, Roan NR. HIV efficiently infects T cells from the endometrium and remodels them to promote systemic viral spread. eLife 2020; 9:55487. [PMID: 32452381 PMCID: PMC7250576 DOI: 10.7554/elife.55487] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
The female reproductive tract (FRT) is the most common site of infection during HIV transmission to women, but viral remodeling complicates characterization of cells targeted for infection. Here, we report extensive phenotypic analyses of HIV-infected endometrial cells by CyTOF, and use a 'nearest neighbor' bioinformatics approach to trace cells to their original pre-infection phenotypes. Like in blood, HIV preferentially targets memory CD4+ T cells in the endometrium, but these cells exhibit unique phenotypes and sustain much higher levels of infection. Genital cell remodeling by HIV includes downregulating TCR complex components and modulating chemokine receptor expression to promote dissemination of infected cells to lymphoid follicles. HIV also upregulates the anti-apoptotic protein BIRC5, which when blocked promotes death of infected endometrial cells. These results suggest that HIV remodels genital T cells to prolong viability and promote viral dissemination and that interfering with these processes might reduce the likelihood of systemic viral spread.
Collapse
Affiliation(s)
- Tongcui Ma
- Gladstone Institute of Virology and Immunology, San Francisco, United States.,Department of Urology, University of California, San Francisco, San Francisco, United States
| | - Xiaoyu Luo
- Gladstone Institute of Virology and Immunology, San Francisco, United States
| | - Ashley F George
- Gladstone Institute of Virology and Immunology, San Francisco, United States.,Department of Urology, University of California, San Francisco, San Francisco, United States
| | - Gourab Mukherjee
- Department of Data Sciences and Operations, University of Southern California, Los Angeles, United States
| | - Nandini Sen
- Departments of Pediatrics and Microbiology and Immunology, Stanford School of Medicine, Stanford, United States
| | - Trimble L Spitzer
- Lt Col, United States AF; Women's Health Clinic, Naval Medical Center, Portsmouth, United States
| | - Linda C Giudice
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, United States
| | - Warner C Greene
- Gladstone Institute of Virology and Immunology, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Nadia R Roan
- Gladstone Institute of Virology and Immunology, San Francisco, United States.,Department of Urology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
4
|
Pérez P, Marín MQ, Lázaro-Frías A, Sorzano CÓS, Gómez CE, Esteban M, García-Arriaza J. Deletion of Vaccinia Virus A40R Gene Improves the Immunogenicity of the HIV-1 Vaccine Candidate MVA-B. Vaccines (Basel) 2020; 8:vaccines8010070. [PMID: 32041218 PMCID: PMC7158668 DOI: 10.3390/vaccines8010070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 02/07/2023] Open
Abstract
Development of a safe and efficacious vaccine against the HIV/AIDS pandemic remains a major scientific goal. We previously described an HIV/AIDS vaccine based on the modified vaccinia virus Ankara (MVA) expressing HIV-1 gp120 and Gag-Pol-Nef (GPN) of clade B (termed MVA-B), which showed moderate immunogenicity in phase I prophylactic and therapeutic clinical trials. Here, to improve the immunogenicity of MVA-B, we generated a novel recombinant virus, MVA-B ΔA40R, by deleting in the MVA-B genome the vaccinia virus (VACV) A40R gene, which encodes a protein with unknown immune function. The innate immune responses triggered by MVA-B ΔA40R in infected human macrophages, in comparison to parental MVA-B, revealed an increase in the mRNA expression levels of interferon (IFN)-β, IFN-induced genes, and chemokines. Compared to priming with DNA-B (a mixture of DNA-gp120 plus DNA-GPN) and boosting with MVA-B, mice immunized with a DNA-B/MVA-B ΔA40R regimen induced higher magnitude of adaptive and memory HIV-1-specific CD4+ and CD8+ T-cell immune responses that were highly polyfunctional, mainly directed against Env. and of an effector memory phenotype, together with enhanced levels of antibodies against HIV-1 gp120. Reintroduction of the A40R gene into the MVA-B ΔA40R genome (virus termed MVA-B ΔA40R-rev) promoted in infected cells high mRNA and protein A40 levels, with A40 protein localized in the cell membrane. MVA-B ΔA40R-rev significantly reduced mRNA levels of IFN-β and of several other innate immune-related genes in infected human macrophages. In immunized mice, MVA-B ΔA40R-rev reduced the magnitude of the HIV-1-specific CD4+ and CD8+ T cell responses compared to MVA-B ΔA40R. These results revealed an immunosuppressive role of the A40 protein, findings relevant for the optimization of poxvirus vectors as vaccines.
Collapse
Affiliation(s)
- Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (P.P.); (M.Q.M.); (A.L.-F.); (C.E.G.); (M.E.)
| | - María Q. Marín
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (P.P.); (M.Q.M.); (A.L.-F.); (C.E.G.); (M.E.)
| | - Adrián Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (P.P.); (M.Q.M.); (A.L.-F.); (C.E.G.); (M.E.)
| | - Carlos Óscar S. Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain;
| | - Carmen E. Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (P.P.); (M.Q.M.); (A.L.-F.); (C.E.G.); (M.E.)
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (P.P.); (M.Q.M.); (A.L.-F.); (C.E.G.); (M.E.)
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (P.P.); (M.Q.M.); (A.L.-F.); (C.E.G.); (M.E.)
- Correspondence: ; Tel.: +34-915-854-560
| |
Collapse
|
5
|
Mouser EEIM, Pollakis G, Smits HH, Thomas J, Yazdanbakhsh M, de Jong EC, Paxton WA. Schistosoma mansoni soluble egg antigen (SEA) and recombinant Omega-1 modulate induced CD4+ T-lymphocyte responses and HIV-1 infection in vitro. PLoS Pathog 2019; 15:e1007924. [PMID: 31487324 PMCID: PMC6728022 DOI: 10.1371/journal.ppat.1007924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/19/2019] [Indexed: 01/17/2023] Open
Abstract
Parasitic helminths evade, skew and dampen human immune responses through numerous mechanisms. Such effects will likely have consequences for HIV-1 transmission and disease progression. Here we analyzed the effects that soluble egg antigen (SEA) from Schistosoma mansoni had on modulating HIV-1 infection and cytokine/chemokine production in vitro. We determined that SEA, specifically through kappa-5, can potently bind to DC-SIGN and thereby blocks DC-SIGN mediated HIV-1 trans-infection (p<0.05) whilst not interfering with cis-infection. DCs exposed to SEA whilst maturing under Th2 promoting conditions, will upon co-culture with naïve T-cells induce a T-cell population that was less susceptible to HIV-1 R5 infection (p<0.05) compared to DCs unexposed to SEA, whereas HIV-1 X4 virus infection was unaffected. This was not observed for DCs exposed to SEA while maturing under Th1 or Th1/Th2 (Tmix) promoting conditions. All T-cell populations induced by SEA exposed DCs demonstrate a reduced capacity to produce IFN-γ and MIP-1β. The infection profile of T-cells infected with HIV-1 R5 was not associated with down-modulation of CCR5 cell surface expression. We further show that DCs maturing under Tmix conditions exposed to plant recombinant omega-1 protein (rω-1), which demonstrates similar functions to natural ω-1, induced T-cell populations that were less sensitive for HIV-1 R5 infection (p<0.05), but not for X4 virus infection. This inhibition associated again with a reduction in IFN-γ and MIP-1β expression, but additionally correlated with reduced CCR5 expression. We have shown that SEA parasite antigens and more specifically rω-1 can modulate HIV-1 infectivity with the potential to influence disease course in co-infected individuals. Parasitic helminths have developed a number of strategies to evade, skew and dampen human immune responses. Such effects will likely have consequences for HIV-1 transmission and disease progression. Here we analyzed the effect that soluble egg antigen (SEA) from Schistosoma mansoni had on HIV-1 infection in vitro. We determined that SEA, through kappa-5, can potently block DC-SIGN mediated HIV-1 trans-infection of CD4+ T-lymphocytes, but not block cis-infection. Dendritic cells (DC) exposed to SEA during maturation under Th2 skewing conditions, induce T-cell populations that are less susceptible to HIV-1 R5 infection compared to cells induced by unexposed DCs. HIV-1 X4 infection was unaffected. This restricted infection profile was not associated with down-modulation of CCR5 surface expression or observed differences in cytokine/chemokine production. Using recombinant omega-1, an abundant component of SEA, HIV-1 R5 infection was similarly inhibited with no effect on HIV-1 X4 infection levels. Hence SEA possesses antigens, namely omega-1, that can modulate HIV-1 infection and potentially influence disease course in co-infected individuals.
Collapse
Affiliation(s)
- Emily EIM Mouser
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Georgios Pollakis
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Hermelijn H. Smits
- Department of Parasitology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Esther C. de Jong
- Department of Cell Biology and Histology, Amsterdam UMC, Location Academic Medical Center, Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, Location Academic Medical Center, Amsterdam, the Netherlands
- * E-mail: (ECdJ); (WAP)
| | - William A. Paxton
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- * E-mail: (ECdJ); (WAP)
| |
Collapse
|
6
|
Abdel-Mohsen M, Kuri-Cervantes L, Grau-Exposito J, Spivak AM, Nell RA, Tomescu C, Vadrevu SK, Giron LB, Serra-Peinado C, Genescà M, Castellví J, Wu G, Del Rio Estrada PM, González-Navarro M, Lynn K, King CT, Vemula S, Cox K, Wan Y, Li Q, Mounzer K, Kostman J, Frank I, Paiardini M, Hazuda D, Reyes-Terán G, Richman D, Howell B, Tebas P, Martinez-Picado J, Planelles V, Buzon MJ, Betts MR, Montaner LJ. CD32 is expressed on cells with transcriptionally active HIV but does not enrich for HIV DNA in resting T cells. Sci Transl Med 2018; 10:eaar6759. [PMID: 29669853 PMCID: PMC6282755 DOI: 10.1126/scitranslmed.aar6759] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/15/2018] [Accepted: 03/27/2018] [Indexed: 12/13/2022]
Abstract
The persistence of HIV reservoirs, including latently infected, resting CD4+ T cells, is the major obstacle to cure HIV infection. CD32a expression was recently reported to mark CD4+ T cells harboring a replication-competent HIV reservoir during antiretroviral therapy (ART) suppression. We aimed to determine whether CD32 expression marks HIV latently or transcriptionally active infected CD4+ T cells. Using peripheral blood and lymphoid tissue of ART-treated HIV+ or SIV+ subjects, we found that most of the circulating memory CD32+ CD4+ T cells expressed markers of activation, including CD69, HLA-DR, CD25, CD38, and Ki67, and bore a TH2 phenotype as defined by CXCR3, CCR4, and CCR6. CD32 expression did not selectively enrich for HIV- or SIV-infected CD4+ T cells in peripheral blood or lymphoid tissue; isolated CD32+ resting CD4+ T cells accounted for less than 3% of the total HIV DNA in CD4+ T cells. Cell-associated HIV DNA and RNA loads in CD4+ T cells positively correlated with the frequency of CD32+ CD69+ CD4+ T cells but not with CD32 expression on resting CD4+ T cells. Using RNA fluorescence in situ hybridization, CD32 coexpression with HIV RNA or p24 was detected after in vitro HIV infection (peripheral blood mononuclear cell and tissue) and in vivo within lymph node tissue from HIV-infected individuals. Together, these results indicate that CD32 is not a marker of resting CD4+ T cells or of enriched HIV DNA-positive cells after ART; rather, CD32 is predominately expressed on a subset of activated CD4+ T cells enriched for transcriptionally active HIV after long-term ART.
Collapse
Affiliation(s)
| | - Leticia Kuri-Cervantes
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Judith Grau-Exposito
- Department of Infectious Diseases, Hospital Universitari Vall d'Hebrón, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Adam M Spivak
- University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Racheal A Nell
- University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | | - Carla Serra-Peinado
- Department of Infectious Diseases, Hospital Universitari Vall d'Hebrón, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Meritxell Genescà
- Department of Infectious Diseases, Hospital Universitari Vall d'Hebrón, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Josep Castellví
- Department of Pathology, Hospital Universitari Vall d´Hebrón, Barcelona 08035, Spain
| | - Guoxin Wu
- Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | | | - Kenneth Lynn
- The Wistar Institute, Philadelphia, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Jonathan Lax Center, Philadelphia FIGHT, Philadelphia, PA 19107, USA
| | | | - Sai Vemula
- Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Kara Cox
- Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Yanmin Wan
- University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Qingsheng Li
- University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Karam Mounzer
- Jonathan Lax Center, Philadelphia FIGHT, Philadelphia, PA 19107, USA
| | - Jay Kostman
- Jonathan Lax Center, Philadelphia FIGHT, Philadelphia, PA 19107, USA
| | - Ian Frank
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | - Gustavo Reyes-Terán
- Instituto Nacional de Enfermedades Respiratorias, Tlalpan 14080, Mexico City, Mexico
| | - Douglas Richman
- Veterans Affairs San Diego Healthcare System and University of California, San Diego, San Diego, CA 92093, USA
| | | | - Pablo Tebas
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Badalona 08916, Barcelona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic 08500, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08908, Catalonia, Spain
| | - Vicente Planelles
- University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Maria J Buzon
- Department of Infectious Diseases, Hospital Universitari Vall d'Hebrón, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain.
| | - Michael R Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
7
|
Abstract
OBJECTIVE The functional polarization of CD4 T cells determines their antimicrobial effector profile, but may also impact the susceptibility to infection with HIV-1. Here, we analyzed the susceptibility of CD4 T cells with different functional polarization to infection with X4 and R5-tropic HIV-1. METHODS CD4 T cells with a Th1, Th2, Th17, and Th9 polarization were subjected to in-vitro infection assays with X4, R5, or vesicular stomatitis virus-G protein-pseudotyped HIV-1. In addition, we sorted differentially polarized CD4 T-cell subsets from individuals treated with antiretroviral therapy and analyzed the tropism of viral env sequences. RESULTS Th9-polarized CD4 T cells and, to a lesser extent, Th2-polarized CD4 T cells expressed higher surface levels of CXCR4, and are more permissive to X4-tropic infection in vitro. In contrast, Th1 and Th17 CD4 T cells exhibited stronger surface expression of CCR5, and were more susceptible to infection with R5-tropic viruses. Correspondingly, the distribution of X4-tropic viral sequences in antiretroviral therapy-treated HIV-1-infected patients was biased toward Th9/Th2 cells, whereas R5-tropic sequences were more frequently observed in Th17 cells. CONCLUSION CD4 T-cell polarization is associated with a distinct susceptibility to X4 and R5-tropic HIV-1 infection.
Collapse
|
8
|
Mitsuki YY, Tuen M, Hioe CE. Differential effects of HIV transmission from monocyte-derived dendritic cells vs. monocytes to IL-17+CD4+ T cells. J Leukoc Biol 2016; 101:339-350. [PMID: 27531931 DOI: 10.1189/jlb.4a0516-216r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/07/2016] [Accepted: 07/26/2016] [Indexed: 12/23/2022] Open
Abstract
HIV infection leads to CD4 helper T cell (Th) loss, but not all Th cells are equally depleted. The contribution of other immune cells in the Th depletion also remains unclear. This study investigates HIV transmission from monocyte-derived dendritic cells (MDDCs) vs. monocytes to Th17 and Th1 cells using an allogeneic coculture model. The addition of HIV to MDDCs increased the expression of the negative regulatory molecule PD-L1 and decreased the expression of the activation markers HLA-DR and CD86, whereas the virus up-regulated HLA-DR and CD86, but not PD-L1, on monocytes. Coculturing of CD4+ T cells with MDDCs pretreated with HIV led to the decline of Th17, but not Th1, responses. In contrast, pretreatment of monocytes with HIV increased Th17 without affecting Th1 responses. The enhanced Th17 responses in the cocultures with HIV-treated monocytes were also accompanied by high numbers of virus-infected CD4+ T cells. The Th17 expansion arose from memory CD4+ T cells with minimal contribution from naïve CD4+ T cells. The Th17-enhancing activity was mediated by the HIV envelope and did not require productive virus infection. Comparison of MDDCs and monocytes further showed that, although HIV-treated MDDCs reduced Th proliferation and increased the activation of the apoptosis mediator caspase-3, HIV-treated monocytes enhanced Th proliferation without increasing the active caspase-3 levels. This study indicates the potential role of distinct myeloid cell populations in shaping Th17 responses during HIV infection.
Collapse
Affiliation(s)
- Yu-Ya Mitsuki
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Tuen
- Veterans Affairs New York Harbor Healthcare System, Manhattan, New York, USA.,Department of Pathology, New York University Langone Medical Center, New York, New York, USA; and
| | - Catarina E Hioe
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; .,James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
9
|
Abdulhaqq SA, Zorrilla C, Kang G, Yin X, Tamayo V, Seaton KE, Joseph J, Garced S, Tomaras GD, Linn KA, Foulkes AS, Azzoni L, VerMilyea M, Coutifaris C, Kossenkov AV, Showe L, Kraiselburd EN, Li Q, Montaner LJ. HIV-1-negative female sex workers sustain high cervical IFNɛ, low immune activation, and low expression of HIV-1-required host genes. Mucosal Immunol 2016; 9:1027-38. [PMID: 26555708 PMCID: PMC4864149 DOI: 10.1038/mi.2015.116] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/30/2015] [Indexed: 02/04/2023]
Abstract
Sex workers practicing in high HIV endemic areas have been extensively targeted to test anti-HIV prophylactic strategies. We hypothesize that in women with high levels of genital exposure to semen changes in cervico-vaginal mucosal and/or systemic immune activation will contribute to a decreased susceptibility to HIV-1 infection. To address this question, we assessed sexual activity and immune activation status (in peripheral blood), as well as cellular infiltrates and gene expression in ectocervical mucosa biopsies in female sex workers (FSWs; n=50), as compared with control women (CG; n=32). FSWs had low-to-absent HIV-1-specific immune responses with significantly lower CD38 expression on circulating CD4(+) or CD8(+) T-cells (both: P<0.001) together with lower cervical gene expression of genes associated with leukocyte homing and chemotaxis. FSWs also had increased levels of interferon-ɛ (IFNɛ) gene and protein expression in the cervical epithelium together with reduced expression of genes associated with HIV-1 integration and replication. A correlative relationship between semen exposure and elevated type-1 IFN expression in FSWs was also established. Overall, our data suggest that long-term condomless sex work can result in multiple changes within the cervico-vaginal compartment that would contribute to sustaining a lower susceptibility for HIV-1 infection in the absence of HIV-specific responses.
Collapse
Affiliation(s)
| | - Carmen Zorrilla
- Maternal-Infant Study Center (CEMI), University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00963
| | - Guobin Kang
- University of Nebraska, School of Biological Sciences and Nebraska Center for Virology, Lincoln, NE 68583
| | | | - Vivian Tamayo
- Maternal-Infant Study Center (CEMI), University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00963
| | - Kelly E. Seaton
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710
| | | | - Sheyla Garced
- Maternal-Infant Study Center (CEMI), University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00963
| | | | - Kristin A. Linn
- Department of Biostatistics and Epidemiology University of Pennsylvania Philadelphia, PA 19104
| | - Andrea S. Foulkes
- University of Massachusetts, Division of Biostatistics and Epidemiology, Amherst, MA 01003
| | | | | | | | | | | | | | - Qingsheng Li
- University of Nebraska, School of Biological Sciences and Nebraska Center for Virology, Lincoln, NE 68583
| | | |
Collapse
|
10
|
Brugia malayi Antigen (BmA) Inhibits HIV-1 Trans-Infection but Neither BmA nor ES-62 Alter HIV-1 Infectivity of DC Induced CD4+ Th-Cells. PLoS One 2016; 11:e0146527. [PMID: 26808476 PMCID: PMC4726616 DOI: 10.1371/journal.pone.0146527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 12/19/2015] [Indexed: 11/19/2022] Open
Abstract
One of the hallmarks of HIV-1 disease is the association of heightened CD4+ T-cell activation with HIV-1 replication. Parasitic helminths including filarial nematodes have evolved numerous and complex mechanisms to skew, dampen and evade human immune responses suggesting that HIV-1 infection may be modulated in co-infected individuals. Here we studied the effects of two filarial nematode products, adult worm antigen from Brugia malayi (BmA) and excretory-secretory product 62 (ES-62) from Acanthocheilonema viteae on HIV-1 infection in vitro. Neither BmA nor ES-62 influenced HIV-1 replication in CD4+ enriched T-cells, with either a CCR5- or CXCR4-using virus. BmA, but not ES-62, had the capacity to bind the C-type lectin dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) thereby inhibiting HIV-1 trans-infection of CD4+ enriched T-cells. As for their effect on DCs, neither BmA nor ES-62 could enhance or inhibit DC maturation as determined by CD83, CD86 and HLA-DR expression, or the production of IL-6, IL-10, IL-12 and TNF-α. As expected, due to the unaltered DC phenotype, no differences were found in CD4+ T helper (Th) cell phenotypes induced by DCs treated with either BmA or ES-62. Moreover, the HIV-1 susceptibility of the Th-cell populations induced by BmA or ES-62 exposed DCs was unaffected for both CCR5- and CXCR4-using HIV-1 viruses. In conclusion, although BmA has the potential capacity to interfere with HIV-1 transmission or initial viral dissemination through preventing the virus from interacting with DCs, no differences in the Th-cell polarizing capacity of DCs exposed to BmA or ES-62 were observed. Neither antigenic source demonstrated beneficial or detrimental effects on the HIV-1 susceptibility of CD4+ Th-cells induced by exposed DCs.
Collapse
|
11
|
García-Díaz D, Rodríguez I, Santisteban Y, Márquez G, Terrero Y, Brown E, Iglesias E. Th2-Th1 shift with the multiantigenic formulation TERAVAC-HIV-1 in Balb/c mice. Immunol Lett 2012. [PMID: 23183092 DOI: 10.1016/j.imlet.2012.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In chronic HIV infection a progressive Th1 to Th2/Th0 cytokine-profile shift is related to disease progression. One of the possible benefits of a therapeutic vaccination might be to counterbalance this phenomenon to allow viral replication control under a Th1-type immune response. TERAVAC-HIV-1 is a multiantigenic formulation vaccine candidate against HIV-1 which comprises the recombinant protein CR3 that contains T cell epitopes and the surface and nucleocapsid antigens of Hepatitis B Virus (HBV). Previous studies showed that such virus like particles of the HBV provide a Th1 adjuvant effect. The present studies examined the capacity of TERAVAC to elicit a Th1 response in the presence of an ongoing HIV-specific Th2-type response in Balb/c mice. To examine this issue, we injected subcutaneously the animals with CR3 or viral lysate in alum which resulted in a Th2-type response. The CR3-specific Th2-type response was verified by induction of IL-4 and IL-10 secretion in ex vivo stimulated splenocytes without secretion of IFN-γ and IgG2a antibodies in serum. Further subcutaneous and simultaneous subcutaneous-nasal immunizations of the same mice with TERAVAC promoted IFN-γ secretion and production of IgG2a antibodies in accordance with a Th1-type response. This result suggests a therapeutic benefit of this vaccine candidate in the restoration of the Th1-type HIV-specific cellular response in seropositive patients.
Collapse
Affiliation(s)
- Darien García-Díaz
- Centro de Ingeniería Genética y Biotecnología (CIGB), P.O. Box 6162, Havana 10600, Cuba
| | | | | | | | | | | | | |
Collapse
|
12
|
Zhang M, Clausell A, Robinson T, Yin J, Chen E, Johnson L, Weiss G, Sabbaj S, Lowe RM, Wagner FH, Goepfert PA, Kutsch O, Cron RQ. Host factor transcriptional regulation contributes to preferential expression of HIV type 1 in IL-4-producing CD4 T cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:2746-57. [PMID: 22875803 DOI: 10.4049/jimmunol.1103129] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
HIV type 1 (HIV-1) replicates preferentially in IL-4-producing CD4 T cells for unclear reasons. We show increased HIV-1 expression is irrespective of viral tropism for chemokine receptors as previously suggested, but rather transcription of the HIV-1 long terminal repeat (LTR) is increased in IL-4-producing CD4 T cells. Increased expression of HIV-1 message is also confirmed in IL-4-producing CD4 T cells from HIV-1-infected individuals ex vivo. In exploring a transcriptional mechanism, we identify a novel c-maf (required for IL-4 expression) transcription factor binding site just upstream of the dual NF-κB/NFAT binding sites in the proximal HIV-1 LTR. We demonstrate that c-maf binds this site in vivo and synergistically augments HIV-1 transcription in cooperation with NFAT2 and NF-κB p65, but not NFAT1 or NF-κB p50. Conversely, small interfering RNA inhibition of c-maf reduces HIV-1 transcription in IL-4-producing T cells. Thus, c-maf increases HIV-1 expression in IL-4-producing CD4 T cells by binding the proximal HIV-1 LTR and augmenting HIV-1 transcription in partnership with NFAT2 and NF-κB p65 specifically. This has important implications for selective targeting of transcription factors during HIV-1 infection because, over the course of HIV-1 progression/AIDS, IL-4-producing T cells frequently predominate and substantially contribute to disease pathology.
Collapse
Affiliation(s)
- Mingce Zhang
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Bi S, Hong PW, Lee B, Baum LG. Galectin-9 binding to cell surface protein disulfide isomerase regulates the redox environment to enhance T-cell migration and HIV entry. Proc Natl Acad Sci U S A 2011; 108:10650-5. [PMID: 21670307 PMCID: PMC3127870 DOI: 10.1073/pnas.1017954108] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Interaction of cell surface glycoproteins with endogenous lectins on the cell surface regulates formation and maintenance of plasma membrane domains, clusters signaling complexes, and controls the residency time of glycoproteins on the plasma membrane. Galectin-9 is a soluble, secreted lectin that binds to glycoprotein receptors to form galectin-glycoprotein lattices on the cell surface. Whereas galectin-9 binding to specific glycoprotein receptors induces death of CD4 Th1 cells, CD4 Th2 cells are resistant to galectin-9 death due to alternative glycosylation. On Th2 cells, galectin-9 binds cell surface protein disulfide isomerase (PDI), increasing retention of PDI on the cell surface and altering the redox status at the plasma membrane. Cell surface PDI regulates integrin function on platelets and also enhances susceptibility of T cells to infection with HIV. We find that galectin-9 binding to PDI on Th2 cells results in increased cell migration through extracellular matrix via β3 integrins, identifying a unique mechanism to regulate T-cell migration. In addition, galectin-9 binding to PDI on T cells potentiates infection with HIV. We identify a mechanism for regulating cell surface redox status via a galectin-glycoprotein lattice, to regulate distinct T-cell functions.
Collapse
Affiliation(s)
- Shuguang Bi
- Departments of Pathology and Laboratory Medicine and
| | - Patrick W. Hong
- Microbiology, Immunology, and Molecular Genetics, University of California School of Medicine, Los Angeles, CA 90095
| | - Benhur Lee
- Departments of Pathology and Laboratory Medicine and
- Microbiology, Immunology, and Molecular Genetics, University of California School of Medicine, Los Angeles, CA 90095
| | - Linda G. Baum
- Departments of Pathology and Laboratory Medicine and
| |
Collapse
|
14
|
Tilton JC, Amrine-Madsen H, Miamidian JL, Kitrinos KM, Pfaff J, Demarest JF, Ray N, Jeffrey JL, Labranche CC, Doms RW. HIV type 1 from a patient with baseline resistance to CCR5 antagonists uses drug-bound receptor for entry. AIDS Res Hum Retroviruses 2010; 26:13-24. [PMID: 20055594 DOI: 10.1089/aid.2009.0132] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CCR5 antagonists are a new class of antiretroviral drugs that block viral entry by disrupting interactions between the viral envelope (Env) glycoprotein and coreceptor. During the CCR100136 (EPIC) Phase IIb study of the CCR5 antagonist aplaviroc (APL) in treatment-naive individuals, a patient was identified who harbored virus strains that exhibited partial resistance to APL at the time of virologic failure. Retrospectively, it was found that APL resistance was present at baseline as well. To investigate the mechanism of APL resistance in this patient, we cloned HIV-1 env genes from plasma obtained at baseline and after virologic failure. Approximately 85% of cloned Envs were functional, and all exhibited partial resistance to APL. All Envs were R5-tropic, were partially resistant to other CCR5 antagonists including maraviroc on cells with high CCR5 expression, but remained sensitive to the fusion inhibitor enfuvirtide. Competition studies with natural CCR5 ligands revealed that the mechanism of drug resistance entailed the use of the drug-bound conformation of CCR5 by the Env proteins obtained from this individual. The degree of drug resistance varied between Env clones, and also varied depending on the cell line used or the donor from whom the primary T cells were obtained. Thus, both virus and host factors contribute to CCR5 antagonist resistance. This study shows that R5 HIV-1 strains resistant to CCR5 inhibitors can arise in patients, confirming a mechanism of resistance previously characterized in vitro. In addition, some patients can harbor CCR5 antagonist-resistant viruses prior to treatment, which may have implications for the clinical use of this new class of antiretrovirals.
Collapse
Affiliation(s)
- John C. Tilton
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Heather Amrine-Madsen
- Departments of Virology and Computational Biology, GlaxoSmithKline, Research Triangle Park, North Carolina 27709
| | - John L. Miamidian
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kathryn M. Kitrinos
- Departments of Virology and Computational Biology, GlaxoSmithKline, Research Triangle Park, North Carolina 27709
- Current address: Kathryn M. Kitrinos, Gilead Sciences Inc., Durham, North Carolina 27707
| | - Jennifer Pfaff
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - James F. Demarest
- Departments of Virology and Computational Biology, GlaxoSmithKline, Research Triangle Park, North Carolina 27709
| | - Neelanjana Ray
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Jerry L. Jeffrey
- Departments of Virology and Computational Biology, GlaxoSmithKline, Research Triangle Park, North Carolina 27709
| | - Celia C. Labranche
- Departments of Virology and Computational Biology, GlaxoSmithKline, Research Triangle Park, North Carolina 27709
- Current address: Celia C. LaBranche, Global Vaccines, Inc., Research Triangle Park, North Carolina 27709
| | - Robert W. Doms
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
15
|
Gosselin A, Monteiro P, Chomont N, Diaz-Griffero F, Said EA, Fonseca S, Wacleche V, El-Far M, Boulassel MR, Routy JP, Sekaly RP, Ancuta P. Peripheral blood CCR4+CCR6+ and CXCR3+CCR6+CD4+ T cells are highly permissive to HIV-1 infection. THE JOURNAL OF IMMUNOLOGY 2009; 184:1604-16. [PMID: 20042588 DOI: 10.4049/jimmunol.0903058] [Citation(s) in RCA: 250] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is limited knowledge on the identity of primary CD4(+) T cell subsets selectively targeted by HIV-1 in vivo. In this study, we established a link between HIV permissiveness, phenotype/homing potential, and lineage commitment in primary CD4(+) T cells. CCR4(+)CCR6(+), CCR4(+)CCR6(-), CXCR3(+)CCR6(+), and CXCR3(+)CCR6(-) T cells expressed cytokines and transcription factors specific for Th17, Th2, Th1Th17, and Th1 lineages, respectively. CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells expressed the HIV coreceptors CCR5 and CXCR4 and were permissive to R5 and X4 HIV replication. CCR4(+)CCR6(-) T cells expressed CXCR4 but not CCR5 and were permissive to X4 HIV only. CXCR3(+)CCR6(-) T cells expressed CCR5 and CXCR4 but were relatively resistant to R5 and X4 HIV in vitro. Total CCR6(+) T cells compared with CCR6(-) T cells harbored higher levels of integrated HIV DNA in treatment-naive HIV-infected subjects. The frequency of total CCR6(+) T cells and those of CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells were diminished in chronically infected HIV-positive subjects, despite viral-suppressive therapy. A high-throughput analysis of cytokine profiles identified CXCR3(+)CCR6(+) T cells as a major source of TNF-alpha and CCL20 and demonstrated a decreased TNF-alpha/IL-10 ratio in CXCR3(+)CCR6(-) T cells. Finally, CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells exhibited gut- and lymph node-homing potential. Thus, we identified CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells as highly permissive to HIV replication, with potential to infiltrate and recruit more CCR6(+) T cells into anatomic sites of viral replication. It is necessary that new therapeutic strategies against HIV interfere with viral replication/persistence in discrete CCR6(+) T cell subsets.
Collapse
Affiliation(s)
- Annie Gosselin
- Department of Microbiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Vetter ML, Johnson ME, Antons AK, Unutmaz D, D'Aquila RT. Differences in APOBEC3G expression in CD4+ T helper lymphocyte subtypes modulate HIV-1 infectivity. PLoS Pathog 2009; 5:e1000292. [PMID: 19197360 PMCID: PMC2631133 DOI: 10.1371/journal.ppat.1000292] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 01/08/2009] [Indexed: 01/01/2023] Open
Abstract
The cytidine deaminases APOBEC3G and APOBEC3F exert anti–HIV-1 activity that is countered by the HIV-1 vif protein. Based on potential transcription factor binding sites in their putative promoters, we hypothesized that expression of APOBEC3G and APOBEC3F would vary with T helper lymphocyte differentiation. Naive CD4+ T lymphocytes were differentiated to T helper type 1 (Th1) and 2 (Th2) effector cells by expression of transcription factors Tbet and GATA3, respectively, as well as by cytokine polarization. APOBEC3G and APOBEC3F RNA levels, and APOBEC3G protein levels, were higher in Th1 than in Th2 cells. T cell receptor stimulation further increased APOBEC3G and APOBEC3F expression in Tbet- and control-transduced, but not in GATA3-transduced, cells. Neutralizing anti–interferon-γ antibodies reduced both basal and T cell receptor-stimulated APOBEC3G and APOBEC3F expression in Tbet- and control-transduced cells. HIV-1 produced from Th1 cells had more virion APOBEC3G, and decreased infectivity, compared to virions produced from Th2 cells. These differences between Th1- and Th2-produced virions were greater for viruses lacking functional vif, but also seen with vif-positive viruses. Over-expression of APOBEC3G in Th2 cells decreased the infectivity of virions produced from Th2 cells, and reduction of APOBEC3G in Th1 cells increased infectivity of virions produced from Th1 cells, consistent with a causal role for APOBEC3G in the infectivity difference. These results indicate that APOBEC3G and APOBEC3F levels vary physiologically during CD4+ T lymphocyte differentiation, that interferon-γ contributes to this modulation, and that this physiological regulation can cause changes in infectivity of progeny virions, even in the presence of HIV-1 vif. Some host cell proteins can hinder, or restrict, the life cycle of HIV-1. APOBEC3G and APOBEC3F are cellular enzymes that decrease HIV-1's ability to replicate in a subsequent target cell if they are present in the virus particle. As a countermeasure, HIV-1 virion infectivity factor (vif) induces degradation of APOBEC3G and APOBEC3F, thereby preventing them from getting into the budding virus. Although vif-defective viruses cannot evade the antiviral effect of APOBEC3G, such viruses are very rarely present in HIV-1-infected humans. It is not yet known whether physiological variation in APOBEC3G and APOBEC3F expression in CD4+ T lymphocytes is substantial enough to reduce vif-positive HIV-1 infectivity. In this study, we found that T helper type 1 (Th1) cells, a subtype of CD4+ lymphocytes, expressed greater amounts of APOBEC3G and APOBEC3F than T helper type 2 (Th2) cells. This difference led to a difference in infectivity of HIV-1 produced from the two cell types, whether vif was expressed or not. These results demonstrate that physiological regulation of APOBEC3G does restrict vif-positive HIV-1, as well as vif-negative HIV-1. In addition, this study reveals biological factors regulating expression of these proteins that may be exploitable for new therapeutic or preventive strategies against HIV-1.
Collapse
Affiliation(s)
- Michael L. Vetter
- Department of Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Megan E. Johnson
- Department of Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Amanda K. Antons
- Department of Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Derya Unutmaz
- Department of Microbiology, New York University, New York, New York, United States of America
| | - Richard T. D'Aquila
- Department of Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
17
|
Pello OM, Martínez-Muñoz L, Parrillas V, Serrano A, Rodríguez-Frade JM, Toro MJ, Lucas P, Monterrubio M, Martínez-A C, Mellado M. Ligand stabilization of CXCR4/delta-opioid receptor heterodimers reveals a mechanism for immune response regulation. Eur J Immunol 2008; 38:537-49. [PMID: 18200497 DOI: 10.1002/eji.200737630] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The CXCR4 chemokine receptor and the delta opioid receptor (DOR) are pertussis toxin-sensitive G protein-coupled receptors (GPCR). Both are widely distributed in brain tissues and immune cells, and have key roles in inflammation processes and in pain sensation on proximal nerve endings. We show that in immune cells expressing CXCR4 and DOR, simultaneous addition of their ligands CXCL12 and [D-Pen2, D-Pen5]enkephalin does not trigger receptor function. This treatment does not affect ligand binding or receptor expression, nor does it promote heterologous desensitization. Our data indicate that CXCR4 and DOR form heterodimeric complexes that are dynamically regulated by the ligands. This is compatible with a model in which GPCR oligomerization leads to suppression of signaling, promoting a dominant negative effect. Knockdown of CXCR4 and DOR signaling by heterodimerization might have repercussions on physiological and pathological processes such as inflammation, pain sensation and HIV-1 infection.
Collapse
Affiliation(s)
- Oscar M Pello
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
In many regions of the world, both schistosomiasis and HIV/AIDS are endemic, resulting in patients harbouring co-infections. Because interaction with host CD4(+) T cells is a characteristic of schistosome as well as HIV-1 infections, bi-directional disease effects may be sufficiently different from sequelae caused by either infectious agent alone to warrant alteration of public health approaches in areas of co-endemnicity. Studies published over the past decade provide useful insights into interactions between schistosomiasis and infection with HIV-1, and overall support the hypothesis that special emphasis on treatment of schistosomiasis in populations with elevated prevalence or risk of HIV-1 infection is justified.
Collapse
Affiliation(s)
- W E Secor
- Division of Parasitic Diseases, Centers for Disease Control and Prevention, Public Health Service, Department of Health and Human Services, Atlanta, Georgia, USA.
| |
Collapse
|
19
|
Kamal SM, El Sayed Khalifa K. Immune modulation by helminthic infections: worms and viral infections. Parasite Immunol 2006; 28:483-96. [PMID: 16965284 DOI: 10.1111/j.1365-3024.2006.00909.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helminthic infections occur worldwide, especially in developing countries. About one-quarter of the world's population, 1.5 billion, are infected with one or more of the major soil-transmitted helminths, including hookworms, ascarids, and whipworms. Schistosomes infect more than 200 million people worldwide with 600 million at risk in 74 countries. The interaction between helminths and the host's immune system provokes particular immunomodulatory and immunoregulatory mechanisms that ensure their survival in the host for years. However, these changes might impair the immunological response to bystander bacterial, viral, and protozoal pathogens and to vaccination. Modulation of the immune system by infection with helminthic parasites is proposed to reduce the levels of allergic responses and to protect against inflammatory bowel disease. In this review, we summarize the immunological milieu associated with helminthic infections and its impact on viral infections, mainly hepatitis B virus, hepatitis C virus, and human immunodeficiency virus in humans and experimental animals.
Collapse
Affiliation(s)
- S M Kamal
- Department of Tropical Medicine, Ain Shams Faculty of Medicine, Heliopolis, Cairo, Egypt.
| | | |
Collapse
|
20
|
Groot F, van Capel TMM, Schuitemaker JHN, Berkhout B, de Jong EC. Differential susceptibility of naïve, central memory and effector memory T cells to dendritic cell-mediated HIV-1 transmission. Retrovirology 2006; 3:52. [PMID: 16916447 PMCID: PMC1562438 DOI: 10.1186/1742-4690-3-52] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Accepted: 08/17/2006] [Indexed: 02/02/2023] Open
Abstract
Background Dendritic cells (DC) have been proposed to facilitate sexual transmission of HIV-1 by capture of the virus in the mucosa and subsequent transmission to CD4+ T cells. Several T cell subsets can be identified in humans: naïve T cells (TN) that initiate an immune response to new antigens, and memory T cells that respond to previously encountered pathogens. The memory T cell pool comprises central memory (TCM) and effector memory cells (TEM), which are characterized by distinct homing and effector functions. The TEM cell subset, which can be further divided into effector Th1 and Th2 cells, has been shown to be the prime target for viral replication after HIV-1 infection, and is abundantly present in mucosal tissues. Results We determined the susceptibility of TN, TCM and TEM cells to DC-mediated HIV-1 transmission and found that co-receptor expression on the respective T cell subsets is a decisive factor for transmission. Accordingly, CCR5-using (R5) HIV-1 was most efficiently transmitted to TEM cells, and CXCR4-using (X4) HIV-1 was preferentially transmitted to TN cells. Conclusion The highly efficient R5 transfer to TEM cells suggests that mucosal T cells are an important target for DC-mediated transmission. This may contribute to the initial burst of virus replication that is observed in these cells. TN cells, which are the prime target for DC-mediated X4 virus transmission in our study, are considered to inefficiently support HIV-1 replication. Our results thus indicate that DC may play a decisive role in the susceptibility of TN cells to X4 tropic HIV-1.
Collapse
Affiliation(s)
- Fedde Groot
- Dept. of Human Retrovirology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Dept. of Cell Biology and Histology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Toni MM van Capel
- Dept. of Cell Biology and Histology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Joost HN Schuitemaker
- Dept. of Cell Biology and Histology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Ben Berkhout
- Dept. of Human Retrovirology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther C de Jong
- Dept. of Cell Biology and Histology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Gallagher M, Malhotra I, Mungai PL, Wamachi AN, Kioko JM, Ouma JH, Muchiri E, King CL. The effects of maternal helminth and malaria infections on mother-to-child HIV transmission. AIDS 2005; 19:1849-55. [PMID: 16227793 DOI: 10.1097/01.aids.0000189846.90946.5d] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate the effect of helminth and/or malaria infection on the risk of HIV infection in pregnant women and its transmission to their offspring. DESIGN A retrospective cohort study of pregnant Kenyan women and their offspring from term, uncomplicated vaginal deliveries (n = 936) with a nested case-control study. METHODS We determined the presence of HIV, malaria, schistosomiasis, lymphatic filariasis, and intestinal helminthes in mothers and tested for HIV antibodies in 12-24 month-old offspring of HIV-positive women. We related these findings to the presence of cord blood lymphocyte activation and cytokine production in response to helminth antigens. RESULTS HIV-positive women (n = 83, 8.9% of all women tested) were 2-fold more likely to have peripheral blood and/or placental malaria (P < 0.025) and a 2.1-fold greater likelihood of lymphatic filariasis infection (P < 0.001) compared to location-and-parity matched HIV-negative women. Women with HIV and malaria tended to show an increased risk for mother-to-child-transmission (MTCT) of HIV, although this difference was not significant. MTCT of HIV, however, was significantly higher in women co-infected with one or more helminthes (48%) verses women without helminth infections (10%, P < 0.01; adjusted odds ratio, 7.3; 95% confidence interval, 2.4-33.7). This increased risk for MTCT of HIV correlated with cord blood lymphocytes production of interleukin-5/interleukin-13 in response to helminth antigens (P < 0.001). CONCLUSION Helminth co-infection is associated with increased risk for MTCT of HIV, possibly by a mechanism in which parasite antigens activates lymphocytes in utero. Treatment of helminthic infections during pregnancy may reduce the risk of MTCT of HIV.
Collapse
Affiliation(s)
- Maureen Gallagher
- Center for Global Health and Diseases and Center for AIDS Research, Case Western Reserve University, Cleveland, OH 44106-7286, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The advent of potent combination antiretroviral therapy has been an important breakthrough in the treatment of HIV-1 infection, resulting in marked reductions in HIV-1-related morbidity and mortality. Antiretroviral therapy has also provided researchers with a powerful tool to perturb the equilibrium of viral production and viral clearance, allowing them to dissect the underlying dynamics that control the pathogenesis of AIDS. Here, we review our current understanding of the sources of HIV-1 production, the estimates for the virion and the host-cell half-lives, and the pathways of virion trafficking and clearance. We also discuss the obstacles that result from the ability of HIV-1 to remain dormant for a prolonged period of time in a subset of long-lived cells, despite an apparently effective antiretroviral treatment.
Collapse
Affiliation(s)
- Viviana Simon
- Aaron Diamond AIDS Research Center, Rockefeller University, 455 First Avenue, New York, New York 10016, USA
| | | |
Collapse
|
23
|
Secor WE, Shah A, Mwinzi PMN, Ndenga BA, Watta CO, Karanja DMS. Increased density of human immunodeficiency virus type 1 coreceptors CCR5 and CXCR4 on the surfaces of CD4(+) T cells and monocytes of patients with Schistosoma mansoni infection. Infect Immun 2003; 71:6668-71. [PMID: 14573694 PMCID: PMC219584 DOI: 10.1128/iai.71.11.6668-6671.2003] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Distribution of chemokine receptors CCR5 and CXCR4, which are also coreceptors for human immunodeficiency virus type 1 invasion of cells, was measured on the surfaces of CD4(+) T cells and monocytes in peripheral blood samples from a group of Kenyan car washers. Patients with active schistosomiasis displayed higher cell surface densities of these receptors than did cured schistosomiasis patients.
Collapse
Affiliation(s)
- W Evan Secor
- Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Public Health Service, U.S. Department of Health and Human Services, Atlanta, Georgia 30341, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Chehimi J, Luo Q, Azzoni L, Shawver L, Ngoubilly N, June R, Jerandi G, Farabaugh M, Montaner LJ. HIV-1 transmission and cytokine-induced expression of DC-SIGN in human monocyte-derived macrophages. J Leukoc Biol 2003; 74:757-63. [PMID: 12960240 DOI: 10.1189/jlb.0503231] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) has been described as an attachment molecule for human immunodeficiency virus type 1 (HIV-1) with the potential to mediate its transmission. We examined DC-SIGN expression in monocyte-derived macrophages (MDM) and its role in viral transmission when MDM were exposed to interleukin (IL)-13, IL-4, or interferon-gamma (IFN-gamma). We show that IL-13 and IL-4 increase transcripts, total protein, and cell-surface expression of DC-SIGN in all MDM tested, IFN-gamma results ranged from no change to up-regulation of surface expression, and message and total protein were, respectively, induced in all and 86% of donors tested. Transmission experiments of HIV-1 X4 between cytokine-treated MDM to Sup-T1 cells showed no association between total transmission and DC-SIGN up-regulation. IL-4 but not IL-13 resulted in a less than twofold increase in MDM viral transmission to CD4+ T cells in spite of a fourfold up-regulation in DC-SIGN expression by either cytokine. In contrast, IFN-gamma treatment induced a decrease in total transmission by at least two-thirds, despite its induction of DC-SIGN. Soluble mannan resulted in a greater inhibition of viral transmission to CD4+ T cells than neutralizing anti-DC-SIGN monoclonal antibody (67-75% vs. 39-48%), supporting the role of mannose-binding receptors in viral transmission. Taken together, results show that DC-SIGN regulation in MDM does not singly predict the transmission potential of this cell type.
Collapse
Affiliation(s)
- Jihed Chehimi
- HIV Immunopathogenesis Laboratory, The Wistar Institute, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vicenzi E, Panina‐Bodignon P, Vallanti G, Di Lucia P, Poli G. Restricted replication of primary HIV‐1 isolates using both CCR5 and CXCR4 in Th2 but not in Th1 CD4
+
T cells. J Leukoc Biol 2002. [DOI: 10.1189/jlb.72.5.913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Elisa Vicenzi
- AIDS Immunopathogenesis Unit, San Raffaele Scientific Institute, Milano, Italy; and
| | | | - Giuliana Vallanti
- AIDS Immunopathogenesis Unit, San Raffaele Scientific Institute, Milano, Italy; and
| | | | - Guido Poli
- AIDS Immunopathogenesis Unit, San Raffaele Scientific Institute, Milano, Italy; and
| |
Collapse
|
26
|
Abstract
Great progress has been made with respect to our understanding of the immunopathogenesis of AIDS and the infectious agent, HIV, that causes the disease. HIV, a human retrovirus with tropism for CD4(+) T cells and monocytes, induces a decrease of T-cell counts, T-cell dysfunction, and, ultimately, immunodeficiency. HIV also causes B-cell dysfunction characterized by polyclonal activation, hypergammaglobulinemia, and lack of specific antibody responses. Chemokine receptors-mainly CCR5 and CXCR4-have been found to be necessary for viral entry into the host cell, a step that can be inhibited by chemokine-related molecules that are ligands for those receptors. After HIV infection, a strong cellular immunity develops and partially controls viral replication. It can take several years for HIV infection to become clinically evident. Studies in long-term nonprogressors have shown the determinant roles of both helper and cytotoxic T cells in the control of HIV disease. Advances in HIV immunology research are currently being applied in the development of prophylactic and therapeutic vaccines.
Collapse
Affiliation(s)
- Javier Chinen
- Allergy and Immunology Program, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|