1
|
Calado M, Pires D, Conceição C, Santos-Costa Q, Anes E, Azevedo-Pereira JM. Human immunodeficiency virus transmission-Mechanisms underlying the cell-to-cell spread of human immunodeficiency virus. Rev Med Virol 2023; 33:e2480. [PMID: 37698498 DOI: 10.1002/rmv.2480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/25/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023]
Abstract
Despite the success of combined antiretroviral therapy in controlling viral load and reducing the risk of human immunodeficiency virus (HIV) transmission, an estimated 1.5 million new infections occurred worldwide in 2021. These new infections are mainly the result of sexual intercourse and thus involve cells present on the genital mucosa, such as dendritic cells (DCs), macrophages (Mø) and CD4+ T lymphocytes. Understanding the mechanisms by which HIV interacts with these cells and how HIV exploits these interactions to establish infection in a new human host is critical to the development of strategies to prevent and control HIV transmission. In this review, we explore how HIV has evolved to manipulate some of the physiological roles of these cells, thereby gaining access to strategic cellular niches that are critical for the spread and pathogenesis of HIV infection. The interaction of HIV with DCs, Mø and CD4+ T lymphocytes, and the role of the intercellular transfer of viral particles through the establishment of the infectious or virological synapses, but also through membrane protrusions such as filopodia and tunnelling nanotubes (TNTs), and cell fusion or cell engulfment processes are presented and discussed.
Collapse
Affiliation(s)
- Marta Calado
- Faculty of Pharmacy, Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - David Pires
- Faculty of Pharmacy, Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Rio de Mouro, Portugal
| | - Carolina Conceição
- Faculty of Pharmacy, Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Quirina Santos-Costa
- Faculty of Pharmacy, Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Elsa Anes
- Faculty of Pharmacy, Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - José Miguel Azevedo-Pereira
- Faculty of Pharmacy, Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
2
|
Wang W, Truong K, Ye C, Sharma S, He H, Liu L, Wen M, Misra A, Zhou P, Kimata JT. Engineered CD4 T cells expressing a membrane anchored viral inhibitor restrict HIV-1 through cis and trans mechanisms. Front Immunol 2023; 14:1167965. [PMID: 37781368 PMCID: PMC10538569 DOI: 10.3389/fimmu.2023.1167965] [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: 02/17/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023] Open
Abstract
HIV-1 infection of target cells can occur through either cell-free virions or cell-cell transmission in a virological synapse, with the latter mechanism of infection reported to be 100- to 1,000-fold more efficient. Neutralizing antibodies and entry inhibitors effectively block cell-free HIV-1, but with few exceptions, they display much less inhibitory activity against cell-mediated HIV-1 transmission. Previously, we showed that engineering HIV-1 target cells by genetically linking single-chain variable fragments (scFvs) of antibodies to glycosyl phosphatidylinositol (GPI) potently blocks infection by cell-free virions and cell-mediated infection by immature dendritic cell (iDC)-captured HIV-1. Expression of scFvs on CD4+ cell lines by transduction with X5 derived anti-HIV-1 Env antibody linked to a GPI attachment signal directs GPI-anchored scFvs into lipid rafts of the plasma membrane. In this study, we further characterize the effect of GPI-scFv X5 on cell-cell HIV-1 transmission from DCs to target cells. We report that expression of GPI-scFv X5 in transduced CD4+ cell lines and human primary CD4+ T cells potently restricts viral replication in iDC- or mDC-captured HIV-1 in trans. Using live-cell imaging, we observed that when GPI-GFP or GPI-scFv X5 transduced T cells are co-cultured with iDCs, GPI-anchored proteins enrich in contact zones and subsequently migrate from T cells into DCs, suggesting that transferred GPI-scFv X5 interferes with HIV-1 infection of iDCs. We conclude that GPI-scFv X5 on the surface of transduced CD4+ T cells not only potently blocks cell-mediated infection by DCs, but it transfers from transduced cells to the surface of iDCs and neutralizes HIV-1 replication in iDCs. Our findings have important implications for HIV-1 antibody-based immunotherapies as they demonstrate a viral inhibitory effect that extends beyond the transduced CD4+ T cells to iDCs which can enhance HIV-1 replication.
Collapse
Affiliation(s)
- Weiming Wang
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Khanghy Truong
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Chaobaihui Ye
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Suman Sharma
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Huan He
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Lihong Liu
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Michael Wen
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Anisha Misra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Paul Zhou
- Unit of Anti-Viral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
3
|
Valdebenito S, Ono A, Rong L, Eugenin EA. The role of tunneling nanotubes during early stages of HIV infection and reactivation: implications in HIV cure. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:169-186. [PMID: 37476291 PMCID: PMC10355284 DOI: 10.1515/nipt-2022-0015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/30/2022] [Indexed: 07/22/2023]
Abstract
Tunneling nanotubes (TNTs), also called cytonemes or tumor microtubes, correspond to cellular processes that enable long-range communication. TNTs are plasma membrane extensions that form tubular processes that connect the cytoplasm of two or more cells. TNTs are mostly expressed during the early stages of development and poorly expressed in adulthood. However, in disease conditions such as stroke, cancer, and viral infections such as HIV, TNTs proliferate, but their role is poorly understood. TNTs function has been associated with signaling coordination, organelle sharing, and the transfer of infectious agents such as HIV. Here, we describe the critical role and function of TNTs during HIV infection and reactivation, as well as the use of TNTs for cure strategies.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Akira Ono
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Libin Rong
- Department of Mathematics, University of Florida, Gainesville, FL, USA
| | - Eliseo A. Eugenin
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| |
Collapse
|
4
|
Caputo V, Libera M, Sisti S, Giuliani B, Diotti RA, Criscuolo E. The initial interplay between HIV and mucosal innate immunity. Front Immunol 2023; 14:1104423. [PMID: 36798134 PMCID: PMC9927018 DOI: 10.3389/fimmu.2023.1104423] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
Human Immunodeficiency Virus (HIV) is still one of the major global health issues, and despite significant efforts that have been put into studying the pathogenesis of HIV infection, several aspects need to be clarified, including how innate immunity acts in different anatomical compartments. Given the nature of HIV as a sexually transmitted disease, one of the aspects that demands particular attention is the mucosal innate immune response. Given this scenario, we focused our attention on the interplay between HIV and mucosal innate response: the different mucosae act as a physical barrier, whose integrity can be compromised by the infection, and the virus-cell interaction induces the innate immune response. In addition, we explored the role of the mucosal microbiota in facilitating or preventing HIV infection and highlighted how its changes could influence the development of several opportunistic infections. Although recent progress, a proper characterization of mucosal innate immune response and microbiota is still missing, and further studies are needed to understand how they can be helpful for the formulation of an effective vaccine.
Collapse
|
5
|
Baratella M, Iannone V, Cavarelli M, Foglieni C, Viganò P, Moog C, Elmore U, Nozza S, Alfano M, Salonia A, Dispinseri S, Scarlatti G. Human seminal plasma stimulates the migration of CD11c+ mononuclear phagocytes to the apical side of the colonic epithelium without altering the junctional complexes in an ex vivo human intestinal model. Front Immunol 2023; 14:1133886. [PMID: 37033941 PMCID: PMC10073423 DOI: 10.3389/fimmu.2023.1133886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Human immunodeficiency virus type 1 (HIV) transmission mostly occurs through the genital and intestinal mucosae. Although HIV-1 transmission has been extensively investigated, gaps remain in understanding the initial steps of HIV entry through the colonic mucosa. We previously showed that HIV can selectively trigger mononuclear phagocytes (MNP) to migrate within colonic epithelial cells to sample virions. Mucosal exposure to human seminal plasma (HSP), rich in pro- and anti-inflammatory cytokines, chemokines and growth factors, may as well induce alterations of the colonic mucosa and recruit immune cells, hence, affecting pathogen sampling and transmission. Methods Here, we studied the role of HSP on the paracellular intestinal permeability by analyzing the distribution of two proteins known to play a key role in controlling the intestinal barrier integrity, namely the tight junctions-associated junctional adhesion molecule (JAM-A) and the adherents junction associated protein E-cadherin (E-CAD), by immunofluorescence and confocal microscopy. Also, we evaluated if HSP promotes the recruitment of MNP cells, specifically, the CD11c and CD64 positive MNPs, to the apical side of the human colonic mucosa. At this scope, HSP of HIV-infected and uninfected individuals with known fertility status was tested for cytokines, chemokines and growth factors concentration and used in an ex vivo polarized colonic tissue culture system to mimic as closely as possible the physiological process. Results HSP showed statistically significant differences in cytokines and chemokines concentrations between the three groups of donors, i.e. HIV infected, or uninfected fertile or randomly identified. Nevertheless, we showed that in the ex vivo tissue culture HSP in general, neither affected the morphological structure of the colonic mucosa nor modulated the paracellular intestinal permeability. Interestingly, CD11c+ MNP cells migrated to the apical surface of the colonic epithelium regardless, if incubated with HIV-infected or -uninfected HSPs, while CD64+ MNP cells, did not change their distribution within the colonic mucosa. Discussion In conclusion, even if HSP did not perturb the integrity of the human colonic mucosa, it affected the migration of a specific subset of MNPs that express CD11c towards the apical side of the colonic mucosa, which in turn may be involved in pathogen sampling.
Collapse
Affiliation(s)
- Marco Baratella
- Viral Evolution and Transmission Group, Division of Immunology, Transplantation, and Infectious Diseases, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
- *Correspondence: Marco Baratella,
| | - Valeria Iannone
- Viral Evolution and Transmission Group, Division of Immunology, Transplantation, and Infectious Diseases, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Mariangela Cavarelli
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Université Paris-Saclay, Inserm, Paris, France
| | - Chiara Foglieni
- Cardiovascular Research Center, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Paola Viganò
- Reproductive Sciences Laboratory, Gynecology/Obstetrics Unit, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Christiane Moog
- INSERM U1109, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Ugo Elmore
- Department of Gastrointestinal Surgery, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Silvia Nozza
- Division of Infectious Diseases, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology, Unit of Urology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Andrea Salonia
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology, Unit of Urology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Stefania Dispinseri
- Viral Evolution and Transmission Group, Division of Immunology, Transplantation, and Infectious Diseases, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Group, Division of Immunology, Transplantation, and Infectious Diseases, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
6
|
Wang T, Gu Y, Ran L, Tan X, Peng S. Ways of HIV transmission in China: The effect of age, period, and cohort. Front Public Health 2022; 10:941941. [PMID: 36159288 PMCID: PMC9493025 DOI: 10.3389/fpubh.2022.941941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/09/2022] [Indexed: 01/25/2023] Open
Abstract
Background Acquired immunodeficiency syndrome (AIDS) is a global pandemic caused by human immunodeficiency virus (HIV), which is transmitted through human behaviors, such as sexual intercourse, intravenous drug injection, and blood transfusion. Rare studies have focused on the evaluation of the effects of culture, society, and HIV-related policies in adjusting people's HIV-related behaviors, i.e., ways of HIV transmission. Methods By taking the new HIV infections in Hubei Province each year from 1995 to 2020 as the sample, our study used the Hierarchical Age-Period-Cohort (HAPC) model to analyze the effects of age, period, and cohort on the trends of ways of HIV transmission. Results From 1995 to 2020, the number of new HIV infections in Hubei presented a general upward trend. A total of 34,636 HIV infections were reported during this period. According to the statistics of the new HIV infections in Hubei Province between 1995 and 2020, there is a negative correlation between age (-0.099, p < 0.001), squared age (-0.002, p < 0.001), and the rate of blood transmission. While there is a positive correlation between age (0.143, p < 0.001), squared age (0.002, p < 0.001), and the rate of HIV infection through sexual transmission. The significant period and cohort effects on ways of HIV transmission were also observed in the Chinese population. Conclusion Sexual and blood transmission are the two main ways of HIV infection in China and Hubei. The trend of blood transmission is in accordance with the wave of blood trade in the early 1990s in China. The trend of sexual transmission indicates an increasing need to promote safer sexual behavior among the older population and later generations and design more tailored, innovative, and diverse HIV prevention strategies, especially for the high-risk groups.
Collapse
Affiliation(s)
- Tang Wang
- School of Public Health, Wuhan University, Wuhan, China
| | - Yaohua Gu
- School of Nursing, Wuhan University, Wuhan, China
| | - Li Ran
- School of Public Health, Wuhan University, Wuhan, China
| | - Xiaodong Tan
- School of Public Health, Wuhan University, Wuhan, China,*Correspondence: Xiaodong Tan
| | - Shuzhen Peng
- Department of Health Management, Renmin Hospital in Huangpi District, Wuhan, China,Shuzhen Peng
| |
Collapse
|
7
|
Delgado-Diaz DJ, Jesaveluk B, Hayward JA, Tyssen D, Alisoltani A, Potgieter M, Bell L, Ross E, Iranzadeh A, Allali I, Dabee S, Barnabas S, Gamieldien H, Blackburn JM, Mulder N, Smith SB, Edwards VL, Burgener AD, Bekker LG, Ravel J, Passmore JAS, Masson L, Hearps AC, Tachedjian G. Lactic acid from vaginal microbiota enhances cervicovaginal epithelial barrier integrity by promoting tight junction protein expression. MICROBIOME 2022; 10:141. [PMID: 36045402 PMCID: PMC9429363 DOI: 10.1186/s40168-022-01337-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/30/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Women with a cervicovaginal microbiota dominated by Lactobacillus spp. are at reduced risk of acquiring sexually transmitted infections including HIV, but the biological mechanisms involved remain poorly defined. Here, we performed metaproteomics on vaginal swab samples from young South African women (n = 113) and transcriptomics analysis of cervicovaginal epithelial cell cultures to examine the ability of lactic acid, a metabolite produced by cervicovaginal lactobacilli, to modulate genital epithelial barrier function. RESULTS Compared to women with Lactobacillus-depleted microbiota, women dominated by vaginal lactobacilli exhibit higher abundance of bacterial lactate dehydrogenase, a key enzyme responsible for lactic acid production, which is independently associated with an increased abundance of epithelial barrier proteins. Physiological concentrations of lactic acid enhance epithelial cell culture barrier integrity and increase intercellular junctional molecule expression. CONCLUSIONS These findings reveal a novel ability of vaginal lactic acid to enhance genital epithelial barrier integrity that may help prevent invasion by sexually transmitted pathogens. Video abstract.
Collapse
Affiliation(s)
- David Jose Delgado-Diaz
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia
| | - Brianna Jesaveluk
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia
| | - Joshua A Hayward
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia
| | - David Tyssen
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Arghavan Alisoltani
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, 92521, USA
| | - Matthys Potgieter
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Liam Bell
- Centre for Proteomic and Genomic Research, Cape Town, 7925, South Africa
| | - Elizabeth Ross
- Centre for Proteomic and Genomic Research, Cape Town, 7925, South Africa
| | - Arash Iranzadeh
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Imane Allali
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, 1014, Rabat, Morocco
| | - Smritee Dabee
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Shaun Barnabas
- Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, 7505, South Africa
| | - Hoyam Gamieldien
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
| | - Jonathan M Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for Infectious Diseases Research (CIDRI) in Africa Wellcome Trust Centre, University of Cape Town, Cape Town, 7925, South Africa
| | - Steven B Smith
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vonetta L Edwards
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Adam D Burgener
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Obstetrics and Gynecology, University of Manitoba, Winnipeg, Canada
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, 7925, South Africa
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jo-Ann S Passmore
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, 4013, South Africa
- National Health Laboratory Service, Cape Town, 7925, South Africa
| | - Lindi Masson
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, 4013, South Africa
- Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Anna C Hearps
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Gilda Tachedjian
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia.
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
8
|
Brady JM, Phelps M, MacDonald SW, Lam EC, Nitido A, Parsons D, Boutros CL, Deal CE, Garcia-Beltran WF, Tanno S, Natarajan H, Ackerman ME, Vrbanac VD, Balazs AB. Antibody-mediated prevention of vaginal HIV transmission is dictated by IgG subclass in humanized mice. Sci Transl Med 2022; 14:eabn9662. [PMID: 35895834 DOI: 10.1126/scitranslmed.abn9662] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
HIV broadly neutralizing antibodies (bNAbs) are capable of both blocking viral entry and driving innate immune responses against HIV-infected cells through their Fc region. Vaccination or productive infection results in a polyclonal mixture of class-switched immunoglobulin G (IgG) antibodies composed of four subclasses, each encoding distinct Fc regions that differentially engage innate immune functions. Despite evidence that innate immunity contributes to protection, the relative contribution of individual IgG subclasses is unknown. Here, we used vectored immunoprophylaxis in humanized mice to interrogate the efficacy of individual IgG subclasses during prevention of vaginal HIV transmission by VRC07, a potent CD4-binding site-directed bNAb. We find that VRC07 IgG2, which lacks Fc-mediated functionality, exhibited substantially reduced protection in vivo relative to other subclasses. Low concentrations of highly functional VRC07 IgG1 yielded substantial protection against vaginal challenge, suggesting that interventions capable of eliciting modest titers of functional IgG subclasses may provide meaningful benefit against infection.
Collapse
Affiliation(s)
- Jacqueline M Brady
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Meredith Phelps
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Scott W MacDonald
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Evan C Lam
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Adam Nitido
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Dylan Parsons
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Christine L Boutros
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Cailin E Deal
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Wilfredo F Garcia-Beltran
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Serah Tanno
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH 03755, USA
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH 03755, USA.,Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Vladimir D Vrbanac
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Alejandro B Balazs
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| |
Collapse
|
9
|
Lin Q, Deng B, Rui J, Guo SB, Hu Q, Chen Q, Tang C, Zhou L, Zhao Z, Lin S, Zhu Y, Yang M, Wang Y, Xu J, Liu X, Yang T, Li P, Li Z, Luo L, Liu W, Liu C, Huang J, Yao M, Nong M, Nong L, Wu J, Luo N, Chen S, Frutos R, Yang S, Li Q, Cui JA, Chen T. Epidemiological Characteristics and Transmissibility of Human Immunodeficiency Virus in Nanning City, China, 2001-2020. Front Public Health 2022; 9:689575. [PMID: 35004557 PMCID: PMC8733253 DOI: 10.3389/fpubh.2021.689575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/11/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Human immunodeficiency virus (HIV) is a single-stranded RNA virus that can weaken the body's cellular and humoral immunity and is a serious disease without specific drug management and vaccine. This study aimed to evaluate the epidemiologic characteristics and transmissibility of HIV. Methods: Data on HIV follow-up were collected in Nanning City, Guangxi Zhuang Autonomous, China. An HIV transmission dynamics model was built to simulate the transmission of HIV and estimate its transmissibility by comparing the effective reproduction number (Reff) at different stages: the rapid growth period from January 2001 to March 2005, slow growth period from April 2005 to April 2011, and the plateau from May 2011 to December 2019 of HIV in Nanning City. Results: High-risk areas of HIV prevalence in Nanning City were mainly concentrated in suburbs. Furthermore, high-risk groups were those of older age, with lower income, and lower education levels. The Reff in each stage (rapid growth, slow growth, and plateau) were 2.74, 1.62, and 1.15, respectively, which suggests the transmissibility of HIV in Nanning City has declined and prevention and control measures have achieved significant results. Conclusion: Over the past 20 years, the HIV incidence in Nanning has remained at a relatively high level, but its development trend has been curbed. Transmissibility was reduced from 2.74 to 1.15. Therefore, the prevention and treatment measures in Nanning City have achieved significant improvement.
Collapse
Affiliation(s)
- Qian Lin
- Development Planning Office, Guangxi Medical University, Nanning, China
| | - Bin Deng
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Jia Rui
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Song-Bai Guo
- Department of Mathematics and Data Science, School of Science, Beijing University of Civil Engineering and Architecture, Beijing, China
| | - Qingqing Hu
- Division of Public Health, School of Medicine, University of Utah, 201 Presidents Circle, Salt Lake, UT, United States
| | - Qiuping Chen
- Laboratory Intertryp CIRAD/IRD, Université de Montpellier, Montpellier, France.,Department of Medical Insurance Office, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Chi Tang
- Division of Director's Office, Nanning Municipal Health Commission, Nanning, China
| | - Lina Zhou
- Department of Nephrology, The Second Hospital of Xiamen Medical College, Xiamen, China
| | - Zeyu Zhao
- Development Planning Office, Guangxi Medical University, Nanning, China.,Laboratory Intertryp CIRAD/IRD, Université de Montpellier, Montpellier, France
| | - Shengnan Lin
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Yuanzhao Zhu
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Meng Yang
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Yao Wang
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Jingwen Xu
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Xingchun Liu
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Tianlong Yang
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Peihua Li
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Zhuoyang Li
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Li Luo
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Weikang Liu
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Chan Liu
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Jiefeng Huang
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| | - Min Yao
- Department of STD and AIDS Prevention and Treatment, Nanning Center for Disease Control and Prevention, Nanning, China
| | - Mengni Nong
- Department of STD and AIDS Prevention and Treatment, Nanning Center for Disease Control and Prevention, Nanning, China
| | - Liping Nong
- Department of STD and AIDS Prevention and Treatment, Nanning Center for Disease Control and Prevention, Nanning, China
| | - Jinglan Wu
- Department of STD and AIDS Prevention and Treatment, Nanning Center for Disease Control and Prevention, Nanning, China
| | - Na Luo
- Department of STD and AIDS Prevention and Treatment, Nanning Center for Disease Control and Prevention, Nanning, China
| | - Shihai Chen
- Division of Director's Office, Nanning Municipal Health Commission, Nanning, China
| | - Roger Frutos
- Department of Medical Insurance Office, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Shixiong Yang
- Department of STD and AIDS Prevention and Treatment, Nanning Center for Disease Control and Prevention, Nanning, China
| | - Qun Li
- Department of Health Emergency, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing-An Cui
- Department of Mathematics and Data Science, School of Science, Beijing University of Civil Engineering and Architecture, Beijing, China
| | - Tianmu Chen
- Department of Science and Technology, School of Public Health, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, China
| |
Collapse
|
10
|
Tanko RF, Bunjun R, Dabee S, Jaumdally SZ, Onono M, Nair G, Palanee-Phillips T, Harryparsad R, Happel AU, Gamieldien H, Qumbelo Y, Sinkala M, Scoville CW, Heller K, Baeten JM, Bosinger SE, Burgener A, Heffron R, Jaspan HB, Passmore JAS. The Effect Of Contraception On Genital Cytokines In Women Randomized To Copper Intrauterine Device, Intramuscular Depot Medroxyprogesterone Acetate Or Levonorgestrel Implant. J Infect Dis 2022; 226:907-919. [PMID: 35263421 PMCID: PMC9470113 DOI: 10.1093/infdis/jiac084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The ECHO Trial randomized women to intramuscular depot medroxyprogesterone acetate (DMPA-IM), levonorgestrel implant (LNG-implant), or copper intrauterine device (Cu-IUD). In a sub-study of the ECHO Trial, we tested the hypothesis that contraceptives influence genital inflammation by comparing cervicovaginal cytokine changes following contraception initiation. In addition, we compared cytokine profiles in women who acquired HIV (cases) versus those remaining HIV-negative (controls). METHODS Women (n=251) from South Africa and Kenya were included. Twenty-seven cervicovaginal cytokines were measured by Luminex at baseline, 1- and 6-month post-contraceptive initiation. In addition, cytokines were measured pre-seroconversion in HIV cases (n=25) and controls (n=100). RESULTS At 6-months post-contraceptive initiation, women using Cu-IUD had increased concentrations of 25/27 cytokines compared to their respective baseline concentrations. In contrast, women initiating DMPA-IM and LNG-implant did not experience changes in cervicovaginal cytokines. Pre-seroconversion concentrations of IL-1β, IL-6, and TNF-α, previously associated with HIV risk, correlated with increased HIV risk in a logistic regression analysis, although not significantly after correcting for multiple comparisons. Adjusting for contraceptive arm did not alter these results. CONCLUSION Although Cu-IUD use broadly increased cervicovaginal cytokine concentrations at 6-months post-insertion, these inflammatory changes were found not to be a significant driver of HIV risk.
Collapse
Affiliation(s)
- Ramla F Tanko
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa.,The Medical Research Centre, Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Rubina Bunjun
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa
| | | | - Shameem Z Jaumdally
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa
| | | | | | - Thesla Palanee-Phillips
- Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Johannesburg, South Africa
| | - Rushil Harryparsad
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa
| | - Anna-Ursula Happel
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa
| | - Hoyam Gamieldien
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa
| | - Yamkela Qumbelo
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa
| | - Musalula Sinkala
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa
| | | | | | - Jared M Baeten
- University of Washington, Seattle, USA.,Gilead Sciences, Foster City, USA
| | - Steven E Bosinger
- Emory University, Atlanta, USA.,Yerkes National Primate Research Center, Atlanta, USA
| | - Adam Burgener
- Center for Global Health and Diseases, Case Western Reserve University, Ohio, USA.,Department of Obstetrics & Gynecology and Medical Microbiology, University of Manitoba, Canada.,Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Heather B Jaspan
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa.,Seattle Children's Research Institute, Seattle, USA
| | - Jo-Ann S Passmore
- Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, University of Cape Town, South Africa.,NRF-DST CAPRISA Centre of Excellence in HIV Prevention, Durban, South Africa.,National Health Laboratory Service, Cape Town, South Africa
| |
Collapse
|
11
|
Detecting Selection in the HIV-1 Genome during Sexual Transmission Events. Viruses 2022; 14:v14020406. [PMID: 35215999 PMCID: PMC8876189 DOI: 10.3390/v14020406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Little is known about whether and how variation in the HIV-1 genome affects its transmissibility. Assessing which genomic features of HIV-1 are under positive or negative selection during transmission is challenging, because very few virus particles are typically transmitted, and random genetic drift can dilute genetic signals in the recipient virus population. We analyzed 30 transmitter–recipient pairs from the Zurich Primary HIV Infection Study and the Swiss HIV Cohort Study using near full-length HIV-1 genomes. We developed a new statistical test to detect selection during transmission, called Selection Test in Transmission (SeTesT), based on comparing the transmitter and recipient virus population and accounting for the transmission bottleneck. We performed extensive simulations and found that sensitivity of detecting selection during transmission is limited by the strong population bottleneck of few transmitted virions. When pooling individual test results across patients, we found two candidate HIV-1 genomic features for affecting transmission, namely amino acid positions 3 and 18 of Vpu, which were significant before but not after correction for multiple testing. In summary, SeTesT provides a general framework for detecting selection based on genomic sequencing data of transmitted viruses. Our study shows that a higher number of transmitter–recipient pairs is required to improve sensitivity of detecting selection.
Collapse
|
12
|
Plesniarski A, Siddik AB, Su RC. The Microbiome as a Key Regulator of Female Genital Tract Barrier Function. Front Cell Infect Microbiol 2022; 11:790627. [PMID: 34976864 PMCID: PMC8719631 DOI: 10.3389/fcimb.2021.790627] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The microbiome, the collection of microbial species at a site or compartment, has been an underappreciated realm of human health up until the last decade. Mounting evidence suggests the microbiome has a critical role in regulating the female genital tract (FGT) mucosa's function as a barrier against sexually transmitted infections (STIs) and pathogens. In this review, we provide the most recent experimental systems and studies for analyzing the interplay between the microbiome and host cells and soluble factors with an influence on barrier function. Key components, such as microbial diversity, soluble factors secreted by host and microbe, as well as host immune system, all contribute to both the physical and immunologic aspects of the FGT mucosal barrier. Current gaps in what is known about the effects of the microbiome on FGT mucosal barrier function are compared and contrasted with the literature of the gut and respiratory mucosa. This review article presents evidence supporting that the vaginal microbiome, directly and indirectly, contributes to how well the FGT protects against infection.
Collapse
Affiliation(s)
- Andrew Plesniarski
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Abu Bakar Siddik
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Ruey-Chyi Su
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
13
|
Gokavi J, Sadawarte S, Shelke A, Kulkarni-Kale U, Thakar M, Saxena V. Inhibition of miR-155 Promotes TGF-β Mediated Suppression of HIV Release in the Cervical Epithelial Cells. Viruses 2021; 13:v13112266. [PMID: 34835072 PMCID: PMC8624372 DOI: 10.3390/v13112266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
TGF-β has been shown to play a differential role in either restricting or aiding HIV infection in different cell types, however its role in the cervical cells is hitherto undefined. Among females, more than 80% of infections occur through heterosexual contact where cervicovaginal mucosa plays a critical role, however the early events during the establishment of infection at female genital mucosa are poorly understood. We earlier showed that increased TGF-β level has been associated with cervical viral shedding in the HIV infected women, however a causal relationship could not be examined. Therefore, here we first established an in vitro cell-associated model of HIV infection in the cervical epithelial cells (ME-180) and demonstrated that TGF-β plays an important role as a negative regulator of HIV release in the infected cervical epithelial cells. Inhibition of miR-155 upregulated TGF-β signaling and mRNA expression of host restriction factors such as APOBEC-3G, IFI-16 and IFITM-3, while decreased the HIV release in ME-180 cells. To conclude, this is the first study to decipher the complex interplay between TGF-β, miR-155 and HIV release in the cervical epithelial cells. Collectively, our data suggest the plausible role of TGF-β in promoting HIV latency in cervical epithelial cells which needs further investigations.
Collapse
Affiliation(s)
- Jyotsna Gokavi
- Division of Immunology and Serology, Indian Council of Medical Research-National AIDS Research Institute, MIDC, Bhosari, Pune 411026, India; (J.G.); (M.T.)
| | - Sharwari Sadawarte
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India; (S.S.); (A.S.); or (U.K.-K.)
| | - Anant Shelke
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India; (S.S.); (A.S.); or (U.K.-K.)
| | - Urmila Kulkarni-Kale
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India; (S.S.); (A.S.); or (U.K.-K.)
| | - Madhuri Thakar
- Division of Immunology and Serology, Indian Council of Medical Research-National AIDS Research Institute, MIDC, Bhosari, Pune 411026, India; (J.G.); (M.T.)
| | - Vandana Saxena
- Division of Immunology and Serology, Indian Council of Medical Research-National AIDS Research Institute, MIDC, Bhosari, Pune 411026, India; (J.G.); (M.T.)
- Correspondence:
| |
Collapse
|
14
|
Jewanraj J, Ngcapu S, Liebenberg LJP. Semen: A modulator of female genital tract inflammation and a vector for HIV-1 transmission. Am J Reprod Immunol 2021; 86:e13478. [PMID: 34077596 PMCID: PMC9286343 DOI: 10.1111/aji.13478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/07/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
In order to establish productive infection in women, HIV must transverse the vaginal epithelium and gain access to local target cells. Genital inflammation contributes to the availability of HIV susceptible cells at the female genital mucosa and is associated with higher HIV transmission rates in women. Factors that contribute to genital inflammation may subsequently increase the risk of HIV infection in women. Semen is a highly immunomodulatory fluid containing several bioactive molecules with the potential to influence inflammation and immune activation at the female genital tract. In addition to its role as a vector for HIV transmission, semen induces profound mucosal changes to prime the female reproductive tract for conception. Still, most studies of mucosal immunity are conducted in the absence of semen or without considering its immune impact on the female genital tract. This review discusses the various mechanisms by which semen exposure may influence female genital inflammation and highlights the importance of routine screening for semen biomarkers in vaginal specimens to account for its impact on genital inflammation.
Collapse
Affiliation(s)
- Janine Jewanraj
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)DurbanSouth Africa
- Department of Medical MicrobiologyUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)DurbanSouth Africa
- Department of Medical MicrobiologyUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Lenine J. P. Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)DurbanSouth Africa
- Department of Medical MicrobiologyUniversity of KwaZulu‐NatalDurbanSouth Africa
| |
Collapse
|
15
|
Chawuke P, van den Berg N, Fouche G, Maharaj V, Shoko T, Johan van der Westhuizen C, Invernizzi L, Alexandre KB. Lobostemon trigonus (Thunb.) H. Buek, a medicinal plant from South Africa as a potential natural microbicide against HIV-1. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114222. [PMID: 34033901 DOI: 10.1016/j.jep.2021.114222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 05/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE There have been different methods proposed to prevent the sexual transmission of HIV-1 and many of them have centered on the use of anti-retrovirals as microbicides. Given that a large section of the African population still relies on herbal medicine, Lobostemon trigonus (L. trigonus), a traditionally used medicinal plant in South Africa to treat HIV-1 was further investigated for its potential as a natural microbicide to prevent the sexual transmission of HIV-1. METHODS The aerial parts of L. trigonus were oven-dried at 80 °C, ground, extracted with boiling water for 30 min and then filtered. The aqueous extract produced was then bioassayed using different HIV-1 inhibition assays. The active components were purified and chemically profiled using ultra-performance liquid chromatography/quadrupole time-of flight mass spectrometry (UPLC-qTOF-MS). The mechanism of HIV-1 inhibition was determined by fusion arrest assay and time of addition assay. Molecular modelling and molecular dynamic simulations, using Schrödinger, were used to better understand the molecule's mechanism of entry inhibition by evaluating their docking affinity and stability against the gp120 of HIV-1. RESULTS The aqueous extract of this plant had a broad spectrum of activity against different subtypes of the virus; neutralizing subtype A, B and C in the TZM-bl cells, with IC50 values ranging from 0.10 to 7.21 μg/mL. The extract was also inhibitory to the virus induced cytopathic effects in CEM-SS cells with an EC50 of 8.9 μg/mL. In addition, it inhibited infection in peripheral blood mononuclear cells (PBMC) and macrophages with IC50 values of 0.97 and 4.4 μg/mL, respectively. In the presence of vaginal and seminal simulants, and in human semen it retained its inhibitory activity albeit with a decrease in efficiency, by about 3-fold. Studies of the mode of action suggested that the extract blocked HIV-1 attachment to target cells. No toxicity was observed when the Lactobacilli strains, L. acidophilus, L. jensenii, and L. crispatus that populate the female genital tract were cultured in the presence of L. trigonus extract. UPLC-qTOF-MS analyses of the purified fraction of the extract, confirmed the presence of six compounds of which four were identified as rosmarinic acid, salvianolic acids B and C and lithospermic acid. The additional molecular dynamic simulations provided further insight into the entry inhibitory characteristics of salvianolic acid B against the HIV-1 gp120, with a stable pose being found within the CD4 binding site. CONCLUSION The data suggests that the inhibitory effect of L. trigonus may be due to the presence of organic acids which are known to possess anti-HIV-1 properties. The molecules salvianolic acids B and C have been identified for the first time in L. trigonus species. Our study also showed that the L. trigonus extract blocked HIV-1 attachment to target cells, and that it has a broad spectrum of activity against different subtypes of the virus; thus, justifying further investigation as a HIV-1 microbicide.
Collapse
Affiliation(s)
- Phindiwe Chawuke
- Council for Scientific and Industrial Research, Pretoria, South Africa; University of Pretoria, Department of Chemistry, Pretoria, South Africa.
| | | | - Gerda Fouche
- University of Pretoria, Department of Chemistry, Pretoria, South Africa.
| | - Vinesh Maharaj
- University of Pretoria, Department of Chemistry, Pretoria, South Africa.
| | - Tinotenda Shoko
- University of Pretoria, Department of Chemistry, Pretoria, South Africa.
| | | | - Luke Invernizzi
- University of Pretoria, Department of Chemistry, Pretoria, South Africa.
| | | |
Collapse
|
16
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|
17
|
Nijmeijer BM, Langedijk CJM, Geijtenbeek TBH. Mucosal Dendritic Cell Subsets Control HIV-1's Viral Fitness. Annu Rev Virol 2021; 7:385-402. [PMID: 32991263 DOI: 10.1146/annurev-virology-020520-025625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cell (DC) subsets are abundantly present in genital and intestinal mucosal tissue and are among the first innate immune cells that encounter human immunodeficiency virus type 1 (HIV-1) after sexual contact. Although DCs have specific characteristics that greatly enhance HIV-1 transmission, it is becoming evident that most DC subsets also have virus restriction mechanisms that exert selective pressure on the viruses during sexual transmission. In this review we discuss the current concepts of the immediate events following viral exposure at genital mucosal sites that lead to selection of specific HIV-1 variants called transmitted founder (TF) viruses. We highlight the importance of the TF HIV-1 phenotype and the role of different DC subsets in establishing infection. Understanding the biology of HIV-1 transmission will contribute to the design of novel treatment strategies preventing HIV-1 dissemination.
Collapse
Affiliation(s)
- Bernadien M Nijmeijer
- Department of Experimental Immunology, Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Catharina J M Langedijk
- Department of Experimental Immunology, Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|
18
|
Carias AM, Schneider JR, Madden P, Lorenzo-Redondo R, Araínga M, Pegu A, Cianci GC, Maric D, Villinger F, Mascola JR, Veazey RS, Hope TJ. Anatomic Distribution of Intravenously Injected IgG Takes Approximately 1 Week to Achieve Stratum Corneum Saturation in Vaginal Tissues. THE JOURNAL OF IMMUNOLOGY 2021; 207:505-511. [PMID: 34162723 DOI: 10.4049/jimmunol.2100253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/16/2021] [Indexed: 01/13/2023]
Abstract
i.v. injected Abs have demonstrated protection against simian HIV infection in rhesus macaques, paving the way for the Antibody Mediated Prevention trial in which at-risk individuals for HIV received an i.v. infusion of the HIV broadly neutralizing Ab VRC01. However, the time needed for these Abs to fully distribute and elicit protection at mucosal sites is still unknown. In this study, we interrogate how long it takes for Abs to achieve peak anatomical levels at the vaginal surface following i.v. injection. Fluorescently labeled VRC01 and/or Gamunex-C were i.v. injected into 24 female rhesus macaques (Macaca mulatta) with vaginal tissues and plasma acquired up to 2 wk postinjection. We found that Ab delivery to the vaginal mucosa occurs in two phases. The first phase involves delivery to the submucosa, occurring within 24 h and persisting beyond 1 wk. The second phase is the delivery through the stratified squamous epithelium, needing ∼1 wk to saturate the stratum corneum. This study has important implications for the efficacy of immunoprophylaxis targeting pathogens at the mucosa.
Collapse
Affiliation(s)
- Ann M Carias
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jeffrey R Schneider
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL
| | - Patrick Madden
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ramon Lorenzo-Redondo
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mariluz Araínga
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD; and
| | - Gianguido C Cianci
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Danijela Maric
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD; and
| | - Ronald S Veazey
- National Primate Research Center, Tulane University School of Medicine, Covington, LA
| | - Thomas J Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL;
| |
Collapse
|
19
|
Xu XQ, Zhang B, Guo L, Liu Y, Meng FZ, Wang X, Hu WH, Khan AI, Ho WZ. Exosomes Transport Anti-Human Immunodeficiency Virus Factors from Human Cervical Epithelial Cells to Macrophages. J Innate Immun 2021; 13:269-279. [PMID: 34082434 PMCID: PMC8460989 DOI: 10.1159/000514886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 01/28/2021] [Indexed: 11/19/2022] Open
Abstract
The female reproductive tract (FRT) is a major site of HIV sexual transmission. As the outermost layer of cells in the FRT, the human cervical epithelial cells (HCEs) have direct contact with HIV or infected cells. Our early work showed that supernatant (SN) from TLR3-activated HCEs contain the antiviral factors that could potently inhibit HIV replication in macrophages. However, it remains to be determined how HCEs transport the anti-HIV factors to macrophages. This follow-up study examined the role of exosomes in HCE-mediated anti-HIV activity. We found that TLR3 activation of HCEs resulted in the release of exosomes that contained multiple IFN-stimulated genes (ISGs: ISG56, OAS1, MxA, and Mx2) and the HIV restriction microRNAs (miR-28, miR-29 family members, miR-125b, miR-150, miR-382, miR-223, miR-20a, and miR-198). The depletion of exosomes from SN of TLR3-activated HCEs diminished HCE-mediated anti-HIV activity in macrophages, indicating that HCE-derived exosomes are responsible for transporting the antiviral molecules to macrophages. These in vitro findings suggest a novel antiviral mechanism by which HCEs participate in the FRT innate immunity against HIV infection. Further in vivo studies are necessary in order to develop an exosome-based delivery system for prevention and treatment of HIV infection through sexual transmission.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
De Vincentis S, Tartaro G, Rochira V, Santi D. HIV and Sexual Dysfunction in Men. J Clin Med 2021; 10:jcm10051088. [PMID: 33807833 PMCID: PMC7961513 DOI: 10.3390/jcm10051088] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Sexual issues tend to go unaddressed in human immunodeficiency virus (HIV) management, although overt sexual dysfunctions are more prevalent in people living with HIV than uninfected people. Erectile dysfunction is the most frequent sexual problem, with a prevalence of 30–50% even in men <40 years of age, but other issues such as loss of libido and ejaculatory disorders should not be overlooked. Peculiar factors related to HIV infection (e.g., fear of virus transmission, changes in body image, HIV-related comorbidities, HIV distress and stigma), alongside classical factors non-related to HIV, should be considered when approaching sexual problems in HIV patients. For this reason, the diagnostic and therapeutic workout of sexual dysfunction in the context of HIV requires a multidisciplinary approach, involving specialists in both infectious diseases and sexual medicine. This narrative review presents an overview of current knowledge on sexual dysfunction in HIV men, deepening the factors driving and taking part in these issues, providing advice for the clinical approach, and underlining the importance of caring for sexual health to improve the quality of life of HIV patients.
Collapse
Affiliation(s)
- Sara De Vincentis
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, Italy; (S.D.V.); (G.T.); (D.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| | - Giulia Tartaro
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, Italy; (S.D.V.); (G.T.); (D.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| | - Vincenzo Rochira
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, Italy; (S.D.V.); (G.T.); (D.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
- Correspondence: ; Tel.: +39-059-396-2453; Fax: +39-059-396-1335
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, 41126 Modena, Italy; (S.D.V.); (G.T.); (D.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41126 Modena, Italy
| |
Collapse
|
21
|
Saba I, Barat C, Chabaud S, Reyjon N, Leclerc M, Jakubowska W, Orabi H, Lachhab A, Pelletier M, Tremblay MJ, Bolduc S. Immunocompetent Human 3D Organ-Specific Hormone-Responding Vaginal Mucosa Model of HIV-1 Infection. Tissue Eng Part C Methods 2021; 27:152-166. [PMID: 33573474 DOI: 10.1089/ten.tec.2020.0333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The lack of appropriate experimental models often limits our ability to investigate the establishment of infections in specific tissues. To reproduce the structural and spatial organization of vaginal mucosae to study human immunodeficiency virus type-1 (HIV-1) infection, we used the self-assembly technique to bioengineer tridimensional vaginal mucosae using human cells extracted from HIV-1-negative healthy pre- and postmenopausal donors. We produced a stroma, free of exogenous material, that can be adapted to generate near-to-native vaginal tissue with the best complexity obtained with seeded epithelial cells on the organ-specific stroma. The autologous engineered tissues had mechanical properties close to native mucosa and shared similar glycogen production, which declined in reconstructed tissues of the postmenopausal donor. The in vitro-engineered tissues were also rendered immune competent by adding human monocyte-derived macrophages (MDMs) on the epithelium or in the stroma layers. The model was infected with HIV-1, and viral replication and transcytosis were observed when immunocompetent reconstructed vaginal mucosa tissue has incorporated MDMs into the stroma and infected with free HIV-1 green fluorescent protein (GFP) viral particles. These data illustrate a natural permissiveness of immunocompetent untransformed human vaginal mucosae to HIV-1 infection. This model offers a physiological tool to explore viral load, HIV-1 transmission in an environment that may contribute to the virus propagation, and new antiviral treatments in vitro. Impact statement This study introduces an innovative immunocompetent three-dimensional human organ-specific vaginal mucosa free of exogenous material for in vitro modeling of human immunodeficiency virus type-1 (HIV-1) infection. The proposed model is histologically close to native tissue, especially by presenting glycogen accumulation in the epithelium's superficial cells, responsive to estrogen, and able to sustain a monocyte-derived macrophage population infected or not by HIV-1 during ∼2 months.
Collapse
Affiliation(s)
- Ingrid Saba
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Corinne Barat
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Stéphane Chabaud
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Nolan Reyjon
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Maude Leclerc
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Weronika Jakubowska
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Hazem Orabi
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Asmaa Lachhab
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Martin Pelletier
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Michel J Tremblay
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Stéphane Bolduc
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec City, Canada
| |
Collapse
|
22
|
Gulati S, Singh P, Diwan A, Mongia A, Kumar S. Functionalized gold nanoparticles: promising and efficient diagnostic and therapeutic tools for HIV/AIDS. RSC Med Chem 2020; 11:1252-1266. [PMID: 34095839 PMCID: PMC8126886 DOI: 10.1039/d0md00298d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Functionalized gold nanoparticles are recognized as promising vehicles in the diagnosis and treatment of human immunodeficiency virus (HIV) owing to their excellent biocompatibility with biomolecules (like DNA or RNA), their potential for multivalency and their unique optical and structural properties. In this context, this review article focuses on the diverse detection abilities and delivery and uptake methodologies of HIV by targeting genes and proteins using gold nanoparticles on the basis of different shapes and sizes in order to promote its effective expression. In addition, recent trends in gold nanoparticle mediated HIV detection, delivery and uptake and treatment are highlighted considering their cytotoxic effects on healthy human cells.
Collapse
Affiliation(s)
- Shikha Gulati
- Department of Chemistry, Sri Venkateswara College, University of Delhi Delhi-110021 India
| | - Parinita Singh
- Department of Chemistry, Sri Venkateswara College, University of Delhi Delhi-110021 India
| | - Anchita Diwan
- Department of Chemistry, Sri Venkateswara College, University of Delhi Delhi-110021 India
| | - Ayush Mongia
- Department of Chemistry, Sri Venkateswara College, University of Delhi Delhi-110021 India
| | - Sanjay Kumar
- Department of Chemistry, Sri Venkateswara College, University of Delhi Delhi-110021 India
| |
Collapse
|
23
|
Macchione MA, Aristizabal Bedoya D, Figueroa FN, Muñoz-Fernández MÁ, Strumia MC. Nanosystems Applied to HIV Infection: Prevention and Treatments. Int J Mol Sci 2020; 21:E8647. [PMID: 33212766 PMCID: PMC7697905 DOI: 10.3390/ijms21228647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
Sexually-transmitted infections (STIs) are a global health concern worldwide as they cause acute diseases, infertility, and significant mortality. Among the bacterial, viral, and parasitic pathogens that can be sexually transmitted, human immunodeficiency virus (HIV) has caused one of the most important pandemic diseases, which is acquired immune deficiency syndrome (AIDS). 32.7 million people have died from AIDS-related illnesses since the start of the epidemic. Moreover, in 2019, 38 million people were living with HIV worldwide. The need to deal with this viral infection becomes more obvious, because it represents not only a problem for public health, but also a substantial economic problem. In this context, it is necessary to focus efforts on developing methods for prevention, detection and treatment of HIV infections that significantly reduce the number of newly infected people and provide a better quality of life for patients. For several decades, biomedical research has been developed allowing quick solutions through the contribution of effective tools. One of them is the use of polymers as vehicles, drug carrier agents, or as macromolecular prodrugs. Moreover, nanosystems (NSs) play an especially important role in the diagnosis, prevention, and therapy against HIV infection. The purpose of this work is to review recent research into diverse NSs as potential candidates for prevention and treatment of HIV infection. Firstly, this review highlights the advantages of using nanosized structures for these medical applications. Furthermore, we provide an overview of different types of NSs used for preventing or combating HIV infection. Then, we briefly evaluate the most recent developments associated with prevention and treatment alternatives. Additionally, the implications of using different NSs are also addressed.
Collapse
Affiliation(s)
- Micaela A. Macchione
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Av. Haya de la Torre y Av. Medina Allende, Córdoba X5000HUA, Argentina; (M.A.M.); (D.A.B.); (F.N.F.)
- Instituto Académico Pedagógico de Ciencias Humanas, Universidad Nacional de Villa María, Arturo Jauretche 1555, Villa María, Córdoba X5220XAO, Argentina
- Instituto de Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA), CONICET, Av. Velez Sárfield 1611, Córdoba X5000HUA, Argentina
| | - Dariana Aristizabal Bedoya
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Av. Haya de la Torre y Av. Medina Allende, Córdoba X5000HUA, Argentina; (M.A.M.); (D.A.B.); (F.N.F.)
- Instituto de Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA), CONICET, Av. Velez Sárfield 1611, Córdoba X5000HUA, Argentina
| | - Francisco N. Figueroa
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Av. Haya de la Torre y Av. Medina Allende, Córdoba X5000HUA, Argentina; (M.A.M.); (D.A.B.); (F.N.F.)
- Instituto de Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA), CONICET, Av. Velez Sárfield 1611, Córdoba X5000HUA, Argentina
| | - María Ángeles Muñoz-Fernández
- Immunology Section, Laboratorio InmunoBiología Molecular, Instituto Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón (HGUGM), Spanish HIV HGM BioBank, C/Dr. Esquerdo 46, 28007 Madrid, Spain;
- Plataforma de Laboratorio, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28007 Madrid, Spain
| | - Miriam C. Strumia
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Av. Haya de la Torre y Av. Medina Allende, Córdoba X5000HUA, Argentina; (M.A.M.); (D.A.B.); (F.N.F.)
- Instituto de Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA), CONICET, Av. Velez Sárfield 1611, Córdoba X5000HUA, Argentina
| |
Collapse
|
24
|
Tamalet C, Devaux C, Dubourg G, Colson P. Resistance to human immunodeficiency virus infection: a rare but neglected state. Ann N Y Acad Sci 2020; 1485:22-42. [PMID: 33009659 DOI: 10.1111/nyas.14452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/25/2020] [Accepted: 07/07/2020] [Indexed: 11/29/2022]
Abstract
The natural history of human immunodeficiency virus (HIV) infection is well understood. In most individuals sexually exposed to HIV, the risk of becoming infected depends on the viral load and on sexual practices and gender. However, a low percentage of individuals who practice frequent unprotected sexual intercourse with HIV-infected partners remain uninfected. Although the systematic study of these individuals has made it possible to identify HIV resistance factors including protective genetic patterns, such epidemiological situations remain paradoxical and not fully understood. In vitro experiments have demonstrated that peripheral blood mononuclear cells (PBMCs) from HIV-free, unexposed blood donors are not equally susceptible to HIV infection; in addition, PBMCs from highly exposed seronegative individuals are generally resistant to infection by primary HIV clinical isolates. We review the literature on permissiveness of PBMCs from healthy blood donors and uninfected hyperexposed individuals to sustained infection and replication of HIV-1 in vitro. In addition, we focus on recent evidence indicating that the gut microbiota may either contribute to natural resistance to or delay replication of HIV infected individuals.
Collapse
Affiliation(s)
- Catherine Tamalet
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Christian Devaux
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Gregory Dubourg
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Philippe Colson
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| |
Collapse
|
25
|
Abstract
Acquired immunodeficiency syndrome (AIDS) was first reported more than 30 years ago among homosexuals in the United States. The epidemiology of this disease indicates that there are three modes of transmission: Blood, mother-to-child, and sexual contact transmission. The pathogen of AIDS is human immunodeficiency virus (HIV), primarily HIV-1. HIV-1 could not break through the structurally and functionally integral skin, and primarily invades the human body through the mucosa irrespective of their integrity. Therefore, the mucosae are the natural transmission routes for HIV-1. The mucosae involved in HIV-1 transmission include the mucosae of the gastrointestinal tract and the urogenital tract. The risks of HIV-1 transmission vary significantly between mucosal sites and individuals, and are associated with mucosal integrity, abundance of target cells, immune status of the host, commensal microbes, and host genetic background. Many factors are closely related to the barrier function of the mucosa, and studies on their roles in HIV-1 invasion could promote the prevention and control of mucosal transmission of HIV-1.
Collapse
Affiliation(s)
- Gui-Bo Yang
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
26
|
Wand H, Reddy T, Dassaye R, Moodley J, Naidoo S, Ramjee G. Estimating prevalence and incidence of sexually transmitted infections among South African women: Implications of combined impacts of risk factors. Int J STD AIDS 2020; 31:1093-1101. [PMID: 32883173 DOI: 10.1177/0956462420915388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In addition to being the epicentre of the HIV epidemic, South Africa also has the highest burden of sexually transmitted infections (STIs) in the world. Therefore, understanding the most influential risk factors of STIs is a research priority. Using the data from 9948 women who resided in KwaZulu Natal, South Africa, we estimated the population attributable risk to quantify the combined impacts of the most influential factors on STI diagnosis. Overall STI prevalence was 20%, and STI incidence was 15 per 100 person-years. Four factors: age at sexual debut, single/not cohabiting, two or more sex partners and parity <3 were identified as the most influential risk factors for STI prevalence and incidence rates. However, these factors collectively associated with only 51% and 53% of the excess STI prevalence and incidence rates, respectively. These relatively modest impacts provide empirical evidence for the significant impacts of unmeasured factors on STIs. Culturally and socially appropriate prevention programs may be more effective to target those at highest risk of STIs.
Collapse
Affiliation(s)
- Handan Wand
- Kirby Institute, University of New South Wales, Kensington, Australia
| | - Tarylee Reddy
- Biostatistics Unit, South African Medical Research Council, Durban, South Africa
| | - Reshmi Dassaye
- HIV Prevention Unit, South African Medical Research Council, Durban, South Africa
| | - Jothi Moodley
- HIV Prevention Unit, South African Medical Research Council, Durban, South Africa
| | - Sarita Naidoo
- HIV Prevention Unit, South African Medical Research Council, Durban, South Africa
| | - Gita Ramjee
- HIV Prevention Unit, South African Medical Research Council, Durban, South Africa
| |
Collapse
|
27
|
Macharia GN, Yue L, Staller E, Dilernia D, Wilkins D, Song H, McGowan E, King D, Fast P, Imami N, Price MA, Sanders EJ, Hunter E, Gilmour J. Infection with multiple HIV-1 founder variants is associated with lower viral replicative capacity, faster CD4+ T cell decline and increased immune activation during acute infection. PLoS Pathog 2020; 16:e1008853. [PMID: 32886726 PMCID: PMC7498102 DOI: 10.1371/journal.ppat.1008853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/17/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
HIV-1 transmission is associated with a severe bottleneck in which a limited number of variants from a pool of genetically diverse quasispecies establishes infection. The IAVI protocol C cohort of discordant couples, female sex workers, other heterosexuals and men who have sex with men (MSM) present varying risks of HIV infection, diverse HIV-1 subtypes and represent a unique opportunity to characterize transmitted/founder viruses (TF) where disease outcome is known. To identify the TF, the HIV-1 repertoire of 38 MSM participants' samples was sequenced close to transmission (median 21 days post infection, IQR 18-41) and assessment of multivariant infection done. Patient derived gag genes were cloned into an NL4.3 provirus to generate chimeric viruses which were characterized for replicative capacity (RC). Finally, an evaluation of how the TF virus predicted disease progression and modified the immune response at both acute and chronic HIV-1 infection was done. There was higher prevalence of multivariant infection compared with previously described heterosexual cohorts. A link was identified between multivariant infection and replicative capacity conferred by gag, whereby TF gag tended to be of lower replicative capacity in multivariant infection (p = 0.02) suggesting an overall lowering of fitness requirements during infection with multiple variants. Notwithstanding, multivariant infection was associated with rapid CD4+ T cell decline and perturbances in the CD4+ T cell and B cell compartments compared to single variant infection, which were reversible upon control of viremia. Strategies aimed at identifying and mitigating multivariant infection could contribute toward improving HIV-1 prognosis and this may involve strategies that tighten the stringency of the transmission bottleneck such as treatment of STI. Furthermore, the sequences and chimeric viruses help with TF based experimental vaccine immunogen design and can be used in functional assays to probe effective immune responses against TF.
Collapse
Affiliation(s)
- Gladys N. Macharia
- Department of Medicine, Imperial College London, London, United Kingdom
- IAVI Human Immunology Laboratory, London, United Kingdom
| | - Ling Yue
- Emory Vaccine Centre, Yerkes National Primate Research Centre, Emory University, Atlanta, GA, United States of America
| | - Ecco Staller
- Department of Medicine, Imperial College London, London, United Kingdom
- IAVI Human Immunology Laboratory, London, United Kingdom
| | - Dario Dilernia
- Emory Vaccine Centre, Yerkes National Primate Research Centre, Emory University, Atlanta, GA, United States of America
| | - Daniel Wilkins
- Emory Vaccine Centre, Yerkes National Primate Research Centre, Emory University, Atlanta, GA, United States of America
| | - Heeyah Song
- Emory Vaccine Centre, Yerkes National Primate Research Centre, Emory University, Atlanta, GA, United States of America
| | - Edward McGowan
- Department of Medicine, Imperial College London, London, United Kingdom
- IAVI Human Immunology Laboratory, London, United Kingdom
| | - Deborah King
- Department of Medicine, Imperial College London, London, United Kingdom
- IAVI Human Immunology Laboratory, London, United Kingdom
| | - Pat Fast
- IAVI, New York, NY, United States of America
| | - Nesrina Imami
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Matthew A. Price
- IAVI, New York, NY, United States of America
- Department of Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA, United States of America
| | - Eduard J. Sanders
- Kenya Medical Research Institute-Wellcome Trust, Kilifi, Kenya
- Nuffield Department of Clinical Medicine, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Headington, United Kingdom
| | - Eric Hunter
- Emory Vaccine Centre, Yerkes National Primate Research Centre, Emory University, Atlanta, GA, United States of America
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States of America
| | - Jill Gilmour
- Department of Medicine, Imperial College London, London, United Kingdom
- IAVI Human Immunology Laboratory, London, United Kingdom
| |
Collapse
|
28
|
Wand H, Morris N, Dassaye R, Reddy T, Ramjee G. Correlates of Sexually Transmitted Infections Among South African Women Using Individual- and Community-Level Factors: Results from Generalized Additive Mixed Models. ARCHIVES OF SEXUAL BEHAVIOR 2020; 49:1875-1886. [PMID: 30767180 PMCID: PMC6944771 DOI: 10.1007/s10508-018-1315-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/29/2018] [Accepted: 09/21/2018] [Indexed: 06/09/2023]
Abstract
South Africa has the highest burden of human immunodeficiency virus (HIV) infections in the world. There is also growing evidence that an individual's risk of contracting HIV is increased by the presence of other sexually transmitted infections (STIs). The primary objective of this study was to examine the association between the prevalence of STIs in a cohort of South African women who enrolled in HIV prevention trials (2002-2012). The current study linked the individual factors with the community-level characteristics using geo-referencing. These multi-level data were analyzed in generalized additive mixed models settings. In the multivariate logistic regression model, younger age (odds ratio [OR] 4.30, 95% CI 3.20, 5.77 and OR 2.72, 95% CI 2.02, 3.66 for age < 25 and 25-29, respectively); being single/not cohabiting (OR 4.57, 95% CI 3.18, 6.53), two + sex partners (OR 1.46, 95% CI 1.18,1.80); parity < 2 (OR 2.04, 95% CI 1.53, 2.72), parity = 2 (OR 1.85, 95% CI 1.37, 2.48), and using injectables (contraceptive) (OR 1.53, 95% CI 1.13, 2.06) were all significantly associated with increased prevalence of STIs. Women who resided in the communities with high proportions of female headed-households were also significantly at higher risk for STIs (OR 1.20, p = .0025). Because these factors may reflect characteristics of the larger groups who share similar cultural norms and social environments, they can provide considerable insight into the spread of STIs. Prevention strategies based on individual and community-level drivers of STIs are likely to be the most effective means of targeting and reaching those at greatest risk of infection. This strategy has the potential to play a significant role in the epidemic's trajectory.
Collapse
Affiliation(s)
- Handan Wand
- Kirby Institute, University of New South Wales, Kensington, NSW, 2052, Australia.
| | - Natashia Morris
- Biostatistics Unit, South African Medical Research Council, Durban, Kwazulu-Natal, South Africa
| | - Reshmi Dassaye
- HIV Prevention Research Unit, South African Medical Research Council, Westville, KwaZulu-Natal, South Africa
| | - Tarylee Reddy
- Biostatistics Unit, South African Medical Research Council, Durban, Kwazulu-Natal, South Africa
| | - Gita Ramjee
- Biostatistics Unit, South African Medical Research Council, Durban, Kwazulu-Natal, South Africa
| |
Collapse
|
29
|
Agarwal R, Trivedi J, Mitra D. High yield production of recombinant cyanovirin-N (antiviral lectin) exhibiting significant anti-HIV activity, from a rationally selected Escherichia coli strain. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
New GMP manufacturing processes to obtain thermostable HIV-1 gp41 virosomes under solid forms for various mucosal vaccination routes. NPJ Vaccines 2020; 5:41. [PMID: 32435515 PMCID: PMC7235025 DOI: 10.1038/s41541-020-0190-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/28/2020] [Indexed: 01/02/2023] Open
Abstract
The main objective of the MACIVIVA European consortium was to develop new Good Manufacturing Practice pilot lines for manufacturing thermostable vaccines with stabilized antigens on influenza virosomes as enveloped virus-like particles. The HIV-1 gp41-derived antigens anchored in the virosome membrane, along with the adjuvant 3M-052 (TLR7/8 agonist) on the same particle, served as a candidate vaccine for the proof of concept for establishing manufacturing processes, which can be directly applied or adapted to other virosomal vaccines or lipid-based particles. Heat spray-dried powders suitable for nasal or oral delivery, and freeze-dried sublingual tablets were successfully developed as solid dosage forms for mucosal vaccination. The antigenic properties of vaccinal antigens with key gp41 epitopes were maintained, preserving the original immunogenicity of the starting liquid form, and also when solid forms were exposed to high temperature (40 °C) for up to 3 months, with minimal antigen and adjuvant content variation. Virosomes reconstituted from the powder forms remained as free particles with similar size, virosome uptake by antigen-presenting cells in vitro was comparable to virosomes from the liquid form, and the presence of excipients specific to each solid form did not prevent virosome transport to the draining lymph nodes of immunized mice. Virosome integrity was also preserved during exposure to <−15 °C, mimicking accidental freezing conditions. These “ready to use and all-in-one” thermostable needle-free virosomal HIV-1 mucosal vaccines offer the advantage of simplified logistics with a lower dependence on the cold chain during shipments and distribution.
Collapse
|
31
|
Suphaphiphat K, Bernard-Stoecklin S, Gommet C, Delache B, Dereuddre-Bosquet N, Kent SJ, Wines BD, Hogarth PM, Le Grand R, Cavarelli M. Innate and Adaptive Anti-SIV Responses in Macaque Semen: Implications for Infectivity and Risk of Transmission. Front Immunol 2020; 11:850. [PMID: 32528466 PMCID: PMC7247827 DOI: 10.3389/fimmu.2020.00850] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/14/2020] [Indexed: 12/16/2022] Open
Abstract
HIV-1 infection is transmitted primarily by sexual exposure, with semen being the principal contaminated fluid. However, HIV-specific immune response in semen has been understudied. We investigated specific parameters of the innate, cellular, and humoral immune response that may affect semen infectivity in macaques infected with SIVmac251. Serial semen levels of cytokines and chemokines, SIV-specific antibodies, neutralization, and FcγR-mediated functions and SIV-specific T-cell responses were assessed and compared to systemic responses across 53 cynomolgus macaques. SIV infection induced an overall inflammatory state in the semen. Several pro-inflammatory molecules correlated with SIV virus levels. Effector CD8+ T cells were expanded in semen upon infection. SIV-specific CD8+ T-cells that expressed multiple effector molecules (IFN-γ+MIP-1β+TNF+/-) were induced in the semen of a subset of SIV-infected macaques, but this did not correlate with local viral control. SIV-specific IgG, commonly capable of engaging the FcγRIIIa receptor, was detected in most semen samples although this positively correlated with seminal viral load. Several inflammatory immune responses in semen develop in the context of higher levels of SIV seminal plasma viremia. These inflammatory immune responses could play a role in viral transmission and should be considered in the development of preventive and prophylactic vaccines.
Collapse
Affiliation(s)
- Karunasinee Suphaphiphat
- CEA-Université Paris Sud-INSERM U1184, “Immunology of Viral Infections and Auto-Immune Diseases”, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Sibylle Bernard-Stoecklin
- CEA-Université Paris Sud-INSERM U1184, “Immunology of Viral Infections and Auto-Immune Diseases”, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Céline Gommet
- CEA-Université Paris Sud-INSERM U1184, “Immunology of Viral Infections and Auto-Immune Diseases”, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Benoit Delache
- CEA-Université Paris Sud-INSERM U1184, “Immunology of Viral Infections and Auto-Immune Diseases”, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- CEA-Université Paris Sud-INSERM U1184, “Immunology of Viral Infections and Auto-Immune Diseases”, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Stephen J. Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
- ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia
| | - Bruce D. Wines
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - P. Mark Hogarth
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Roger Le Grand
- CEA-Université Paris Sud-INSERM U1184, “Immunology of Viral Infections and Auto-Immune Diseases”, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Mariangela Cavarelli
- CEA-Université Paris Sud-INSERM U1184, “Immunology of Viral Infections and Auto-Immune Diseases”, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| |
Collapse
|
32
|
Matrix Metalloproteinases Expressed in Response to Bacterial Vaginosis Disrupt the Endocervical Epithelium, Increasing Transmigration of HIV. Infect Immun 2020; 88:IAI.00041-20. [PMID: 32094253 DOI: 10.1128/iai.00041-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/01/2020] [Indexed: 02/07/2023] Open
Abstract
Bacterial vaginosis (BV), a disorder of the female reproductive tract (FRT) in which a healthy Lactobacillus-dominant microflora is replaced by BV-associated bacteria (BVAB), can significantly increase the incidence of human immunodeficiency virus (HIV) acquisition. Discerning the effect of BV on the mucosal epithelium of the FRT may yield novel preventatives and therapeutics for HIV infection. Here, we investigated barrier dysfunction of the endocervix by host-derived factors, secreted in response to BV, as a potential cause of HIV infection. Using a polarized endocervical cell culture system, we determined that conditioned media (CM) from endocervical cells cocultured with BVAB (endocervical+BVAB CM), as well as cervicovaginal fluid (CVF) from women with BV, disrupted epithelial polarization. We assessed host matrix metalloproteinases (MMPs) as the BV-associated secreted factors which disrupt the endocervical epithelium. MMPs were overexpressed in endocervical+BVAB CM and CVF from women with BV and were capable of disrupting endocervical epithelial polarization. When we cocultured polarized endocervical cells with HIV-1-infected lymphocyte-derived cells, we discovered endocervical+BVAB CM and MMPs significantly increased the transmigration of virus through the epithelium, and treatment with an MMP inhibitor decreased these effects. When we examined the effect of CVF on HIV-1 transmigration through endocervical epithelium, we demonstrated that CVF samples with greater concentrations of BV-associated MMPs increased viral transmigration. Our results suggest MMPs increase HIV-1 infection by disrupting the endocervical epithelium, permitting transmigration of virus through the epithelium to infect underlying target cells.
Collapse
|
33
|
Alexandre K, Malatji K, Mulaudzi T. Comparison of the antiviral activity of the microbicide candidate griffithsin and its tandemers derivatives against different modes of HIV-1 transmission. Virology 2020; 544:12-20. [PMID: 32174510 DOI: 10.1016/j.virol.2020.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 01/22/2023]
Abstract
Tandemers 2MG, 2MG3, 3MG and 4MG are derivatives of the potent anti-HIV-1 microbicide candidate griffithsin (GRFT). We compared these compounds anti-HIV-1 activity to GRFT using the viruses CAP206.08 and CAAN5342.A2 that have decreased sensitivity to this lectin. The 2MG and 2MG3 tandemers had similar activity to GRFT against cell-free and cell-associated viruses, while 3MG and 4MG were significantly more potent. Furthermore, the restoration of the 234N or 295N glycan in these viruses, known to increase sensitivity to GRFT, also increased sensitivity to 2MG and 2MG3, and not to 3MG and 4MG. In addition, GRFT resistant viruses generated in-vitro were equally resistant to 2MG and 2MG3 while they had considerably low resistance to 3MG and 4MG. Lastly, all five compounds showed increased inhibitory activity in seminal and vaginal simulants although the effect was more pronounced in the former. These data support further studies of tandemers as potential microbicides.
Collapse
Affiliation(s)
- Kabamba Alexandre
- Council for Scientific and Industrial Research, Emerging Research Area Platform, Next Generation Health Cluster, Pretoria, Gauteng, South Africa.
| | - Kanyane Malatji
- Council for Scientific and Industrial Research, Emerging Research Area Platform, Next Generation Health Cluster, Pretoria, Gauteng, South Africa; University of the Witwatersrand, School of Pathology, Johannesburg, South Africa
| | - Takalani Mulaudzi
- Council for Scientific and Industrial Research, Emerging Research Area Platform, Next Generation Health Cluster, Pretoria, Gauteng, South Africa; University of the Witwatersrand, School of Pathology, Johannesburg, South Africa
| |
Collapse
|
34
|
Kalidasan V, Theva Das K. Lessons Learned From Failures and Success Stories of HIV Breakthroughs: Are We Getting Closer to an HIV Cure? Front Microbiol 2020; 11:46. [PMID: 32082282 PMCID: PMC7005723 DOI: 10.3389/fmicb.2020.00046] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/10/2020] [Indexed: 12/19/2022] Open
Abstract
There is a continuous search for an HIV cure as the success of ART in blocking HIV replication and the role of CD4+ T cells in HIV pathogenesis and immunity do not entirely eradicate HIV. The Berlin patient, who is virus-free, serves as the best model for a 'sterilizing cure' and many experts are trying to mimic this approach in other patients. Although failures were reported among Boston and Essen patients, the setbacks have provided valuable lessons to strengthen cure strategies. Following the Berlin patient, two more patients known as London and Düsseldorf patients might be the second and third person to be cured of HIV. In all the cases, the patients underwent chemotherapy regimen due to malignancy and hematopoietic stem cell transplantation (HSCT) which required matching donors for CCR5Δ32 mutation - an approach that may not always be feasible. The emergence of newer technologies, such as long-acting slow-effective release ART (LASER ART) and CRISPR/Cas9 could potentially overcome the barriers due to HIV latency and persistency and eliminate the need for CCR5Δ32 mutation donor. Appreciating the failure and success stories learned from these HIV breakthroughs would provide some insight for future HIV eradication and cure strategies.
Collapse
Affiliation(s)
| | - Kumitaa Theva Das
- Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| |
Collapse
|
35
|
Clark M, Maselko M. Transgene Biocontainment Strategies for Molecular Farming. FRONTIERS IN PLANT SCIENCE 2020; 11:210. [PMID: 32194598 PMCID: PMC7063990 DOI: 10.3389/fpls.2020.00210] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/11/2020] [Indexed: 05/21/2023]
Abstract
Advances in plant synthetic biology promise to introduce novel agricultural products in the near future. 'Molecular farms' will include crops engineered to produce medications, vaccines, biofuels, industrial enzymes, and other high value compounds. These crops have the potential to reduce costs while dramatically increasing scales of synthesis and provide new economic opportunities to farmers. Current transgenic crops may be considered safe given their long-standing use, however, some applications of molecular farming may pose risks to human health and the environment. Unwanted gene flow from engineered crops could potentially contaminate the food supply, and affect wildlife. There is also potential for unwanted gene flow into engineered crops which may alter their ability to produce compounds of interest. Here, we briefly discuss the applications of molecular farming and explore the various genetic and physical methods that can be used for transgene biocontainment. As yet, no technology can be applied to all crop species, such that a combination of approaches may be necessary. Effective biocontainment is needed to enable large scale molecular farming.
Collapse
Affiliation(s)
- Michael Clark
- Applied Biosciences, Macquarie University, North Ryde, NSW, Australia
| | - Maciej Maselko
- Applied Biosciences, Macquarie University, North Ryde, NSW, Australia
- CSIRO Health and Biosecurity, Canberra, ACT, Australia
- CSIRO Synthetic Biology Future Science Platform, Brisbane, QLD, Australia
- *Correspondence: Maciej Maselko,
| |
Collapse
|
36
|
Nijmeijer BM, Geijtenbeek TBH. Negative and Positive Selection Pressure During Sexual Transmission of Transmitted Founder HIV-1. Front Immunol 2019; 10:1599. [PMID: 31354736 PMCID: PMC6635476 DOI: 10.3389/fimmu.2019.01599] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022] Open
Abstract
Sexual transmission of HIV-1 consists of processes that exert either positive or negative selection pressure on the virus. The sum of these selection pressures lead to the transmission of only one specific HIV-1 strain, termed the transmitted founder virus. Different dendritic cell subsets are abundantly present at mucosal sites and, interestingly, these DC subsets exert opposite pressure on viral selection during sexual transmission. In this review we describe receptors and cellular compartments in DCs that are involved in HIV-1 communication leading to either viral restriction by the host or further dissemination to establish a long-lived reservoir. We discuss the current understanding of host antiretroviral restriction factors against HIV-1 and specifically against the HIV-1 transmitted founder virus. We will also discuss potential clinical implications for exploiting these intrinsic restriction factors in developing novel therapeutic targets. A better understanding of these processes might help in developing strategies against HIV-1 infections by targeting dendritic cells.
Collapse
Affiliation(s)
- Bernadien M Nijmeijer
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
37
|
Abstract
Microbicides are antiseptic topical drugs that help directly or indirectly inhibit the penetration of an infectious agent into the human body, thereby preventing the sexual transmission of HIV-infection and other sexually transmitted diseases. Microbicides have an antiviral mechanism of action in the sexual transmission of HIV and affect the components of mucosal immunity in the vagina. In this article, the pharmaceutical and biomedical aspects of microbicide application are examined and diverse classifications of microbicides are presented. For each group of chemicals, the most important representatives and their mechanisms of action are described. This article also presents the structure and function of mucosal immunity, and shows the importance of the mucosal immune response in the sexual transmission of HIV. This work also exhibits the experimental models for testing of candidate microbicides. For each compound described, a review of preclinical research and clinical trials is provided, covering its development as a microbicide. This paper gives an overview of microbicides, a new class of chemically diverse immunobiological medications reducing the risk of sexual transmission of HIV. The use of microbicides is believed to curb the HIV/AIDS epidemic in the nearest future.
Collapse
|
38
|
Quispe Calla NE, Vicetti Miguel RD, Glick ME, Kwiek JJ, Gabriel JM, Cherpes TL. Exogenous oestrogen inhibits genital transmission of cell-associated HIV-1 in DMPA-treated humanized mice. J Int AIDS Soc 2019; 21. [PMID: 29334191 PMCID: PMC5810324 DOI: 10.1002/jia2.25063] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 01/02/2018] [Indexed: 12/11/2022] Open
Abstract
Introduction HIV affects more women than any other life‐threatening infectious agent, and most infections are sexually transmitted. HIV must breach the female genital tract mucosal barrier to establish systemic infection, and clinical studies indicate virus more easily evades this barrier in women using depot‐medroxyprogesterone acetate (DMPA) and other injectable progestins for contraception. Identifying a potential mechanism for this association, we learned DMPA promotes susceptibility of wild‐type mice to genital herpes simplex virus type 2 (HSV‐2) infection by reducing genital tissue expression of the cell‐cell adhesion molecule desmoglein‐1 (DSG‐1) and increasing genital mucosal permeability. Conversely, DMPA‐mediated increases in genital mucosal permeability and HSV‐2 susceptibility were eliminated in mice concomitantly administered exogenous oestrogen (E). To confirm and extend these findings, herein we used humanized mice to define effects of systemic DMPA and intravaginal (ivag) E administration on susceptibility to genital infection with cell‐associated HIV‐1. Methods Effects of DMPA or an intravaginal (ivag) E cream on engraftment of NOD‐scid‐IL‐2Rgcnull (NSG) mice with human peripheral blood mononuclear cells (hPBMCs) were defined with flow cytometry. Confocal microscopy was used to evaluate effects of DMPA, DMPA and E cream, or DMPA and the pharmacologically active component of the cream on vaginal tissue DSG‐1 expression and genital mucosal permeability to low molecular weight (LMW) molecules and hPBMCs. In other studies, hPBMC‐engrafted NSG mice (hPBMC‐NSG) received DMPA or DMPA and ivag E cream before genital inoculation with 106 HIV‐1‐infected hPBMCs. Mice were euthanized 10 days after infection, and plasma HIV‐1 load quantified by qRT‐PCR and splenocytes used to detect HIV‐1 p24 antigen via immunohistochemistry and infectious virus via TZM‐bl luciferase assay. Results Whereas hPBMC engraftment was unaffected by DMPA or E treatment, mice administered DMPA and E (cream or the pharmacologically active cream component) displayed greater vaginal tissue expression of DSG‐1 protein and decreased vaginal mucosal permeability to LMW molecules and hPBMCs versus DMPA‐treated mice. DMPA‐treated hPBMC‐NSG mice were also uniformly susceptible to genital transmission of cell‐associated HIV‐1, while no animal concomitantly administered DMPA and E cream acquired systemic HIV‐1 infection. Conclusion Exogenous E administration reduces susceptibility of DMPA‐treated humanized mice to genital HIV‐1 infection.
Collapse
Affiliation(s)
- Nirk E Quispe Calla
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Melissa E Glick
- The Ohio State University (OSU) College of Veterinary Medicine, Columbus, OH, USA
| | - Jesse J Kwiek
- Department of Microbiology, OSU College of Arts and Sciences, Columbus, OH, USA
| | | | - Thomas L Cherpes
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
39
|
Cherne MD, Hall J, Kellner A, Chong CF, Cole AL, Cole AM. Avirulins, a Novel Class of HIV-1 Reverse Transcriptase Inhibitors Effective in the Female Reproductive Tract Mucosa. Viruses 2019; 11:v11050408. [PMID: 31052477 PMCID: PMC6563246 DOI: 10.3390/v11050408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/25/2022] Open
Abstract
While extensive research efforts have decreased human immunodeficiency virus (HIV) transmissions and mortalities, new challenges have arisen in the fight to eradicate HIV. Drug resistance to antiretroviral therapy threatens infected individuals, while the prevalence of heterosexual transmission creates an urgent need for therapies effective in the female reproductive tract (FRT) mucosa. We screened a library of 2095 small molecule compounds comprising a unique chemical space, purchased from Asinex Corporation, for antiviral activity against human immunodeficiency virus type 1 (HIV-1) strain BaL and identified several molecular representatives of a unique class of HIV-1 inhibitors, which we termed “Avirulins.” We determined that Avirulins were active against clinical isolates of HIV-1 from genetically variant subtypes, several of which have reduced sensitivity to other antivirals. Avirulins displayed specific dose-dependent inhibition of the HIV-1 drug target, reverse transcriptase (RT). Avirulins were effective against several nucleoside RT-inhibitor resistant strains of HIV-1, as well as one nonnucleoside RT-inhibitor resistant strain containing a 106A mutation, suggesting a noncompetitive mechanism of action. Drugs, which are damaging to the FRT, can increase the risk of HIV-1 transmission. We therefore explored the cytotoxicity of Avirulins against epithelial cells derived from the FRT and found no significant toxicity, even at the highest concentrations tested. Importantly, Avirulin antiviral activity was not diminished in human cervico–vaginal fluid, suggesting retained potency in the milieu of the FRT. Based on these promising results, Avirulins should be valuable chemical scaffolds for development into next-generation treatments and preventatives that target HIV-1.
Collapse
Affiliation(s)
- Michelle D Cherne
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| | - Jesse Hall
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| | - Alisha Kellner
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| | - Christine F Chong
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| | - Amy L Cole
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| | - Alexander M Cole
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| |
Collapse
|
40
|
Perez-Zsolt D, Cantero-Pérez J, Erkizia I, Benet S, Pino M, Serra-Peinado C, Hernández-Gallego A, Castellví J, Tapia G, Arnau-Saz V, Garrido J, Tarrats A, Buzón MJ, Martinez-Picado J, Izquierdo-Useros N, Genescà M. Dendritic Cells From the Cervical Mucosa Capture and Transfer HIV-1 via Siglec-1. Front Immunol 2019; 10:825. [PMID: 31114569 PMCID: PMC6503733 DOI: 10.3389/fimmu.2019.00825] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/28/2019] [Indexed: 01/06/2023] Open
Abstract
Antigen presenting cells from the cervical mucosa are thought to amplify incoming HIV-1 and spread infection systemically without being productively infected. Yet, the molecular mechanism at the cervical mucosa underlying this viral transmission pathway remains unknown. Here we identified a subset of HLA-DR+ CD14+ CD11c+ cervical DCs at the lamina propria of the ectocervix and the endocervix that expressed the type-I interferon inducible lectin Siglec-1 (CD169), which promoted viral uptake. In the cervical biopsy of a viremic HIV-1+ patient, Siglec-1+ cells harbored HIV-1-containing compartments, demonstrating that in vivo, these cells trap viruses. Ex vivo, a type-I interferon antiviral environment enhanced viral capture and trans-infection via Siglec-1. Nonetheless, HIV-1 transfer via cervical DCs was effectively prevented with antibodies against Siglec-1. Our findings contribute to decipher how cervical DCs may boost HIV-1 replication and promote systemic viral spread from the cervical mucosa, and highlight the importance of including inhibitors against Siglec-1 in microbicidal strategies.
Collapse
Affiliation(s)
- Daniel Perez-Zsolt
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jon Cantero-Pérez
- Department of Infectious Diseases, Vall d'Hebron Institut de Recerca, Barcelona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | | | - Susana Benet
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Pino
- IrsiCaixa AIDS Research Institute, Badalona, Spain
| | - Carla Serra-Peinado
- Department of Infectious Diseases, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Alba Hernández-Gallego
- Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,Pathology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Josep Castellví
- Pathology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain.,Department of Morphological Sciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gustavo Tapia
- Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,Pathology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Morphological Sciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vicent Arnau-Saz
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Infectious Diseases, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | | | - Antoni Tarrats
- Department of Obstetrics and Gynecology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Maria J Buzón
- Department of Infectious Diseases, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Meritxell Genescà
- Department of Infectious Diseases, Vall d'Hebron Institut de Recerca, Barcelona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
41
|
Abstract
Most new HIV infections, over 80%, occur through sexual transmission. During sexual transmission, the virus must bypass specific female and male reproductive tract anatomical barriers to encounter viable target cells. Understanding the generally efficient ability of these barrier to exclude HIV and the precise mechanisms of HIV translocation beyond these genital barriers is essential for vaccine and novel therapeutic development. In this review, we explore the mucosal, barriers of cervico-vaginal and penile tissues that comprise the female and male reproductive tracts. The unique cellular assemblies f the squamous and columnar epithelium are illustrate highlighting their structure and function. Each anatomical tissue offers a unique barrier to virus entry in healthy individuals. Unfortunately barrier dysfunction can lead to HIV transmission. How these diverse mucosal barriers have the potential to fail is considered, highlighting those anatomical areas that are postulated to offer a weaker barrier and are; therefore, more susceptible to viral ingress. Risk factors, such as sexually transmitted infections, microbiome dysbiosis, and high progestin environments are also associated with increased acquisition of HIV. How these states may affect the integrity of mucosal barriers leading to HIV acquisition are discussed suggesting mechanisms of transmission and revealing potential targets for intervention.
Collapse
Affiliation(s)
- Ann M Carias
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Lurie 9-290, Chicago, IL 60611, USA
| | - Thomas J Hope
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Lurie 9-290, Chicago, IL 60611, USA
| |
Collapse
|
42
|
Sanyal A, Shen C, Ding M, Reinhart TA, Chen Y, Sankapal S, Gupta P. Neisseria gonorrhoeae uses cellular proteins CXCL10 and IL8 to enhance HIV-1 transmission across cervical mucosa. Am J Reprod Immunol 2019; 81:e13111. [PMID: 30903720 PMCID: PMC6540971 DOI: 10.1111/aji.13111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/01/2019] [Accepted: 03/07/2019] [Indexed: 12/14/2022] Open
Abstract
Problem Neisseria gonorrhoeae (NG) infection has been shown to increase sexual transmission of HIV‐1. However, the mechanism of NG‐induced enhanced HIV‐1 transmission is unknown. Methods (a) The cervical tissues were exposed to NG, and cytokine induction was monitored by measuring cytokine proteins in culture supernatants and cytokine mRNAs in tissues. (b) Transcription and replication of HIV‐1 in TZM‐bl, U1, and ACH2 cells were measured by Beta‐Gal activity and p24 proteins in the supernatant, respectively. (c) HIV‐1 transmission was assayed in an organ culture system by measuring transmitted HIV‐1 in supernatant and HIV‐1 gag mRNA in the tissues. (d) Transcriptome analysis was done using second generation sequencing. Results (a) NG induced membrane ruffling of epithelial layer, caused migration of CD3+ cells to the intraepithelial region, and induced high levels of inflammatory cytokines IL‐1β and TNF‐α. (b) NG‐induced supernatants (NGIS) increased HIV‐1 transcription, induced HIV‐1 from latently infected cells, and increased transmission of HIV‐1 across cervical mucosa. (c) Transcriptome analysis of the epithelial layer of the tissues exposed to NG, and HIV‐1 showed significant upregulation of CXCL10 and IL8. IL‐1β increased the induction of CXCL10 and IL‐8 expression in cervical mucosa with a concomitant increase in HIV‐1 transmission. Conclusion We present a model in which IL‐1β produced from cervical epithelium during NG exposure increases CXCL10 and IL8 in epithelia. This in turn causes upon HIV‐1 infection, the migration of HIV‐1 target cells toward the subepithelium, resulting in increased HIV‐1 transcription in the sub‐mucosa and subsequent enhancement of transmission across cervical mucosa.
Collapse
Affiliation(s)
- Anwesha Sanyal
- Department of Infectious Diseases and Microbiology, Pittsburgh, Pennsylvania
| | - Chengli Shen
- Department of Infectious Diseases and Microbiology, Pittsburgh, Pennsylvania
| | - Ming Ding
- Department of Infectious Diseases and Microbiology, Pittsburgh, Pennsylvania
| | | | - Yue Chen
- Department of Infectious Diseases and Microbiology, Pittsburgh, Pennsylvania
| | - Soni Sankapal
- Department of Infectious Diseases and Microbiology, Pittsburgh, Pennsylvania
| | - Phalguni Gupta
- Department of Infectious Diseases and Microbiology, Pittsburgh, Pennsylvania
| |
Collapse
|
43
|
Mesquita L, Galante J, Nunes R, Sarmento B, das Neves J. Pharmaceutical Vehicles for Vaginal and Rectal Administration of Anti-HIV Microbicide Nanosystems. Pharmaceutics 2019; 11:pharmaceutics11030145. [PMID: 30917532 PMCID: PMC6472048 DOI: 10.3390/pharmaceutics11030145] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 12/27/2022] Open
Abstract
Prevention strategies play a key role in the fight against HIV/AIDS. Vaginal and rectal microbicides hold great promise in tackling sexual transmission of HIV-1, but effective and safe products are yet to be approved and made available to those in need. While most efforts have been placed in finding and testing suitable active drug candidates to be used in microbicide development, the last decade also saw considerable advances in the design of adequate carrier systems and formulations that could lead to products presenting enhanced performance in protecting from infection. One strategy demonstrating great potential encompasses the use of nanosystems, either with intrinsic antiviral activity or acting as carriers for promising microbicide drug candidates. Polymeric nanoparticles, in particular, have been shown to be able to enhance mucosal distribution and retention of promising antiretroviral compounds. One important aspect in the development of nanotechnology-based microbicides relates to the design of pharmaceutical vehicles that allow not only convenient vaginal and/or rectal administration, but also preserve or even enhance the performance of nanosystems. In this manuscript, we revise relevant work concerning the selection of vaginal/rectal dosage forms and vehicle formulation development for the administration of microbicide nanosystems. We also pinpoint major gaps in the field and provide pertinent hints for future work.
Collapse
Affiliation(s)
- Letícia Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Joana Galante
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
| | - Rute Nunes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Bruno Sarmento
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, 4585-116 Gandra, Portugal.
| | - José das Neves
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, 4585-116 Gandra, Portugal.
| |
Collapse
|
44
|
CD4-Dependent Modulation of HIV-1 Entry by LY6E. J Virol 2019; 93:JVI.01866-18. [PMID: 30674630 DOI: 10.1128/jvi.01866-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/16/2019] [Indexed: 01/06/2023] Open
Abstract
Lymphocyte antigen 6E (LY6E) is a GPI-anchored, interferon-inducible protein that has been shown to modulate viral infection in a cell type-dependent manner. Our recent work showed that LY6E promotes HIV-1 infection in some high-CD4-expressing cells, including human peripheral blood mononuclear cells (PBMCs) and the SupT1 cell line. In this work, we provide evidence that LY6E inhibits HIV-1 entry and spread in low-CD4-expressing Jurkat cells and human monocyte-derived macrophages (MDMs) through downregulation of the viral receptor CD4. We found that knockdown of LY6E in Jurkat cells and MDMs increases HIV-1 infection, yet overexpression of LY6E in Jurkat cells inhibits HIV-1 entry and replication. LY6E was found to be colocalized with CD4 on the plasma membrane of Jurkat cells and MDMs and enhances CD4 internalization. We artificially manipulated the CD4 level in Jurkat and SupT1 cells and found that overexpression of CD4 in Jurkat cells overcomes the inhibitory effect of LY6E; conversely, blocking the function of CD4 in SupT1 with a neutralizing antibody eliminates the enhancement of LY6E on HIV-1 entry. The CD4-dependent inhibitory phenotype of LY6E in low-CD4-expressing human MDMs can be recapitulated for a panel of transmitted founder viruses and laboratory-adapted HIV-1 strains. Given that HIV-1 can target low-CD4-expressing cells during acute infection yet replicates efficiently in high-CD4-expressing T cells at the late stage of disease, our observation that LY6E differentially modulates HIV-1 replication in a CD4-dependent manner has implications for understanding the complex roles of interferon (IFN)-induced proteins in AIDS pathogenesis.IMPORTANCE The role of IFN-induced genes (ISGs) in viral infection remains incompletely understood. While most ISGs are antiviral, some ISGs have been shown to promote viral infection, including HIV-1 infection. We previously showed that IFN-inducible LY6E protein promotes HIV-1 infection in human PMBCs and high-CD4-expressing SupT1 cells. Here we found that LY6E inhibits HIV-1 entry and replication in low-CD4-expressing MDMs and Jurkat cells. Mechanistically, we demonstrated that LY6E downregulates the cell surface receptor CD4, thus impairing the virus binding to target cells. This is in contrast to the situation of high-CD4-expressing cells, where LY6E predominantly promotes viral membrane fusion. The opposing role of IFN-inducible LY6E in modulating HIV-1 infection highlights the complex roles of ISGs in viral infection and viral pathogenesis.
Collapse
|
45
|
Zalenskaya IA, Chandra N, Yousefieh N, Fang X, Adedipe OE, Jackson SS, Anderson SM, Mauck CK, Schwartz JL, Thurman AR, Doncel GF. Use of contraceptive depot medroxyprogesterone acetate is associated with impaired cervicovaginal mucosal integrity. J Clin Invest 2018; 128:4622-4638. [PMID: 30222141 DOI: 10.1172/jci120583] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Injectable depot medroxyprogesterone acetate (DMPA) is one of the most popular contraception methods in areas of high HIV seroprevalence. Evidence is accumulating that use of DMPA might be associated with an increased risk of HIV-1 acquisition by women; however, mechanisms of this association are not completely understood. The goal of this study was to gain insight into mechanisms underlying the possible link between use of DMPA and risk of HIV-1 acquisition, exploring transcription profiling of ectocervical tissues. METHODS Healthy women received either DMPA (n = 31) or combined oral contraceptive (COC), which has not been linked to an increased risk of HIV acquisition (n = 32). We conducted a comparative microarray-based whole-genome transcriptome profiling of human ectocervical tissues before and after 6 weeks of hormonal contraception use. RESULTS The analysis identified that expression of 235 and 76 genes was significantly altered after DMPA and COC use, respectively. The most striking effect of DMPA, but not COC, was significantly altered expression (mostly downregulation) of many genes strategically involved in the maintenance of mucosal barrier function; the alterations, as indicated by Ingenuity Pathway Analysis (IPA), were most likely due to the DMPA-induced estrogen deficiency. Furthermore, IPA predicted that transcriptome alterations related to ectocervical immune responses were in general compatible with an immunosuppressive effect of DMPA, but, in some women, also with an inflammatory-like response. CONCLUSION Our results suggest that impairment of cervicovaginal mucosal integrity in response to DMPA administration is an important mechanism contributing to the potential increased risk of HIV-1 acquisition in DMPA users. TRIAL REGISTRATION ClinicalTrials.gov NCT01421368. FUNDING This study was supported by the United States Agency for International Development (USAID) under Cooperative Agreement GPO-A-00-08-00005-00.
Collapse
Affiliation(s)
| | - Neelima Chandra
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | - Xi Fang
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | | | | | | | - Jill L Schwartz
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, USA
| | | | - Gustavo F Doncel
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA.,CONRAD, Eastern Virginia Medical School, Arlington, Virginia, USA
| |
Collapse
|
46
|
Shang L, Smith AJ, Duan L, Perkey KE, Wietgrefe S, Zupancic M, Southern PJ, Johnson RP, Carlis JV, Haase AT. Vaccine-Associated Maintenance of Epithelial Integrity Correlated With Protection Against Virus Entry. J Infect Dis 2018; 218:1272-1283. [PMID: 29401315 PMCID: PMC6455945 DOI: 10.1093/infdis/jiy062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
To identify the mechanisms by which human immunodeficiency virus type 1 (HIV-1) might penetrate the epithelial barrier during sexual transmission to women and the mechanisms of vaccine-associated protection against entry, we characterized early epithelial responses to vaginal inoculation of simian immunodeficiency virus strain mac251 (SIVmac251) in naive or SIVmac239Δnef-vaccinated rhesus macaques. Vaginal inoculation induced an early stress response in the cervicovaginal epithelium, which was associated with impaired epithelial integrity, damaged barrier function, and virus and bacterial translocation. In vaccinated animals, early stress responses were suppressed, and the maintenance of epithelial barrier integrity correlated with prevention of virus entry. These vaccine-protective effects were associated with a previously described mucosal system for locally producing and concentrating trimeric gp41 antibodies at the mucosal interface and with formation of SIV-specific immune complexes that block the stress responses via binding to the epithelial receptor FCGR2B and subsequent inhibitory signaling. Thus, blocking virus entry may be one protective mechanism by which locally concentrated non-neutralizing Ab might prevent HIV sexual transmission to women.
Collapse
Affiliation(s)
- L Shang
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - A J Smith
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - L Duan
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - K E Perkey
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - S Wietgrefe
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - M Zupancic
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - P J Southern
- Department of Microbiology and Immunology, Medical School, Minneapolis
| | - R P Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - J V Carlis
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis
| | - A T Haase
- Department of Microbiology and Immunology, Medical School, Minneapolis
| |
Collapse
|
47
|
Takada K, Komine-Aizawa S, Kuramochi T, Ito S, Trinh QD, Pham NTK, Sasano M, Hayakawa S. Lactobacillus crispatus accelerates re-epithelialization in vaginal epithelial cell line MS74. Am J Reprod Immunol 2018; 80:e13027. [PMID: 30144195 DOI: 10.1111/aji.13027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022] Open
Abstract
PROBLEM The functions of vaginal lactobacilli in susceptibility to infectious diseases as regards epithelial barrier integrity and wound healing remain incompletely understood. METHOD OF STUDY Lactobacillus crispatus, one of the most common Lactobacillus species in the vagina and among the most protective against sexually transmitted infections, was cocultured with an immortalized human vaginal epithelial cell line (MS74), and a scratch assay was performed to evaluate re-epithelialization. The concentration of vascular endothelial growth factor A (VEGF) was measured using enzyme-linked immunosorbent assay (ELISA). An immunofluorescence assay was performed to locate the expression of VEGF and VEGF receptor (VEGFR) 1 and 2. The effects of the bacterial supernatant of L. crispatus were also evaluated. RESULTS Lactobacillus crispatus significantly accelerated re-epithelialization of MS74 cells, accompanied by an increase in VEGF concentration. In contrast, heat-killed L. crispatus did not show this effect. The bacterial supernatant of L. crispatus also induced re-epithelialization. The immunoreactivity of VEGF was higher at the scratched edge, whereas VEGFR1 and 2 stained site-independently. Recombinant VEGF induced cell migration in a dose-dependent manner. The bacterial supernatant of L. crispatus also significantly accelerated re-epithelialization in MS74 cells and increased the concentration of VEGF in the culture 24 hours after the scratch. CONCLUSION These results may enhance our knowledge of the importance of L. crispatus in the healing of damaged vaginal epithelium and protection against the consequent risk of pathogenic infections, such as human immunodeficiency virus (HIV), and improve our understanding of vaginal epithelial barrier integrity maintenance by this bacterium.
Collapse
Affiliation(s)
- Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | | | - Shun Ito
- Nihon University School of Medicine, Tokyo, Japan
| | - Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Mari Sasano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.,Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
48
|
Xu XQ, Guo L, Wang X, Liu Y, Liu H, Zhou RH, Gu J, Liu JB, Xu P, Zhou L, Ho WZ. Human Cervical Epithelial Cells Release Antiviral Factors and Inhibit HIV Replication in Macrophages. J Innate Immun 2018; 11:29-40. [PMID: 30032138 DOI: 10.1159/000490586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/03/2018] [Indexed: 12/11/2022] Open
Abstract
The female reproductive tract is a major site of HIV sexual transmission. We here examined whether human cervical epithelial cells (HCEs) can be immunologically activated and produce antiviral factors against HIV. We demonstrated that HCEs (End1/E6E7 cells) possess the functional toll-like receptor (TLR)3 signaling system, which could be activated by Poly I:C and induce multiple cellular HIV restriction factors. The treatment of primary human macrophages with supernatant (SN) from TLR3-activated End1/E6E7 cell cultures resulted in HIV inhibition. This SN-mediated HIV inhibition was mainly through the induction of interferons (IFN)-β and IFN-λs, as the antibodies to IFN-β or IFN-λs receptor could effectively block the SN-mediated anti-HIV effect. Further studies showed that the incubation of macrophages with SN from the activated cervical epithelial cell cultures induced the expression of a number of IFN-stimulated genes (ISGs), including IFN-stimulated gene (ISG15), ISG56, 2', 5'-oligoadenylate synthetase 1 (OAS 1), OAS 2, Myxovirus Resistance A (MxA), MxB, and Guanylate-binding protein 5 (GBP5). In addition, TLR3-activated cells produced the CC chemokines [regulated on activation, normal T cell expressed and secreted (RANTES), Human macrophage inflammatory protein 1 alpha (MIP-1α), MIP-1β] the ligands of HIV entry co-receptor CCR5. These observations support further studies on HCEs as potentially crucial and alternative targets for immunological intervention to control and prevent HIV sexual transmission.
Collapse
Affiliation(s)
- Xi-Qiu Xu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Le Guo
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yu Liu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Hang Liu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Run-Hong Zhou
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Jun Gu
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Jin-Biao Liu
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, China
| | - Pei Xu
- School of Medicine (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Li Zhou
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University School of Medicine, Wuhan,
| | - Wen-Zhe Ho
- Institute of Medical Virology, State Key Laboratory of Virology, Wuhan University School of Basic Medical Sciences, Wuhan, China.,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
49
|
Vaidya NK, Ribeiro RM, Liu P, Haynes BF, Tomaras GD, Perelson AS. Correlation Between Anti-gp41 Antibodies and Virus Infectivity Decay During Primary HIV-1 Infection. Front Microbiol 2018; 9:1326. [PMID: 29973924 PMCID: PMC6019451 DOI: 10.3389/fmicb.2018.01326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022] Open
Abstract
Recent experiments have suggested that the infectivity of simian immunodeficiency virus (SIV) and human immunodeficiency virus type-1 (HIV-1) in plasma decreases over time during primary infection. Because anti-gp41 antibodies are produced early during HIV-1 infection and form antibody-virion complexes, we studied if such early HIV-1 specific antibodies are correlated with the decay in HIV-1 infectivity. Using a viral dynamic model that allows viral infectivity to decay and frequent early viral load data obtained from 6 plasma donors we estimate that HIV-1 infectivity begins to decay after about 2 weeks of infection. The length of this delay is consistent with the time before antibody-virion complexes were detected in the plasma of these donors and is correlated (p = 0.023, r = 0.87) with the time for antibodies to be first detected in plasma. Importantly, we identify that the rate of infectivity decay is significantly correlated with the rate of increase in plasma anti-gp41 IgG concentration (p = 0.046, r = 0.82) and the increase in IgM+IgG anti-gp41 concentration (p = 8.37 × 10−4, r = 0.98). Furthermore, we found that the viral load decay after the peak did not have any significant correlation with the rate of anti-gp41 IgM or IgG increase. These results indicate that early anti-gp41 antibodies may cause viral infectivity decay, but may not contribute significantly to controlling post-peak viral load, likely due to insufficient quantity or affinity. Our findings may be helpful to devise strategies, including antibody-based vaccines, to control acute HIV-1 infection.
Collapse
Affiliation(s)
- Naveen K Vaidya
- Department of Mathematics and Statistics, San Diego State University, San Diego, CA, United States
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics Group, MS K710, Los Alamos National Laboratory, Los Alamos, NM, United States.,Laboratório de Biomatemática, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Pinghuang Liu
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Barton F Haynes
- Duke University School of Medicine, Durham, NC, United States
| | | | - Alan S Perelson
- Theoretical Biology and Biophysics Group, MS K710, Los Alamos National Laboratory, Los Alamos, NM, United States
| |
Collapse
|
50
|
Nazli A, Dizzell S, Zahoor MA, Ferreira VH, Kafka J, Woods MW, Ouellet M, Ashkar AA, Tremblay MJ, Bowdish DM, Kaushic C. Interferon-β induced in female genital epithelium by HIV-1 glycoprotein 120 via Toll-like-receptor 2 pathway acts to protect the mucosal barrier. Cell Mol Immunol 2018; 16:178-194. [PMID: 29553138 PMCID: PMC6355787 DOI: 10.1038/cmi.2017.168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/22/2022] Open
Abstract
More than 40% of HIV infections occur via female reproductive tract (FRT) through heterosexual transmission. Epithelial cells that line the female genital mucosa are the first line of defense against HIV-1 and other sexually transmitted pathogens. These sentient cells recognize and respond to external stimuli by induction of a range of carefully balanced innate immune responses. Previously, we have shown that in response to HIV-1 gp120, the genital epithelial cells (GECs) from upper reproductive tract induce an inflammatory response that may facilitate HIV-1 translocation and infection. In this study, we report that the endometrial and endocervical GECs simultaneously induce biologically active interferon-β (IFNβ) antiviral responses following exposure to HIV-1 that act to protect the epithelial tight junction barrier. The innate antiviral response was directly induced by HIV-1 envelope glycoprotein gp120 and addition of gp120 neutralizing antibody inhibited IFNβ production. Interferon-β was induced by gp120 in upper GECs through Toll-like receptor 2 signaling and required presence of heparan sulfate on epithelial cell surface. The induction of IFNβ was dependent upon activation of transcription factor IRF3 (interferon regulatory factor 3). The IFNβ was biologically active, had a protective effect on epithelial tight junction barrier and was able to inhibit HIV-1 infection in TZM-bl indicator cells and HIV-1 replication in T cells. This is the first report that recognition of HIV-1 by upper GECs leads to induction of innate antiviral pathways. This could explain the overall low infectivity of HIV-1 in the FRT and could be exploited for HIV-1 prophylaxis.
Collapse
Affiliation(s)
- Aisha Nazli
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Sara Dizzell
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Muhammad Atif Zahoor
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Victor H Ferreira
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Jessica Kafka
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Matthew William Woods
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Michel Ouellet
- Department of Medical Biology, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Michel J Tremblay
- Department of Medical Biology, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Dawn Me Bowdish
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada. .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|