51
|
Effectiveness of Xielikang capsules in treating HIV-related diarrhea by increasing the plasma concentration of interleukin-17. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(17)30314-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
52
|
Durand M, Chartrand-Lefebvre C, Baril JG, Trottier S, Trottier B, Harris M, Walmsley S, Conway B, Wong A, Routy JP, Kovacs C, MacPherson PA, Monteith KM, Mansour S, Thanassoulis G, Abrahamowicz M, Zhu Z, Tsoukas C, Ancuta P, Bernard N, Tremblay CL. The Canadian HIV and aging cohort study - determinants of increased risk of cardio-vascular diseases in HIV-infected individuals: rationale and study protocol. BMC Infect Dis 2017; 17:611. [PMID: 28893184 PMCID: PMC5594495 DOI: 10.1186/s12879-017-2692-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/17/2017] [Indexed: 01/08/2023] Open
Abstract
Background With potent antiretroviral drugs, HIV infection is becoming a chronic disease. Emergence of comorbidities, particularly cardiovascular disease (CVD) has become a leading concern for patients living with the infection. We hypothesized that the chronic and persistent inflammation and immune activation associated with HIV disease leads to accelerated aging, characterized by CVD. This will translate into higher incidence rates of CVD in HIV infected participants, when compared to HIV negative participants, after adjustment for traditional CVD risk factors. When characterized further using cardiovascular imaging, biomarkers, immunological and genetic profiles, CVD associated with HIV will show different characteristics compared to CVD in HIV-negative individuals. Methods/design The Canadian HIV and Aging cohort is a prospective, controlled cohort study funded by the Canadian Institutes of Health Research. It will recruit patients living with HIV who are aged 40 years or older or have lived with HIV for 15 years or more. A control population, frequency matched for age, sex, and smoking status, will be recruited from the general population. Patients will attend study visits at baseline, year 1, 2, 5 and 8. At each study visit, data on complete medical and pharmaceutical history will be captured, along with anthropometric measures, a complete physical examination, routine blood tests and electrocardiogram. Consenting participants will also contribute blood samples to a research biobank. The primary outcome is incidence of a composite of: myocardial infarction, coronary revascularization, stroke, hospitalization for angina or congestive heart failure, revascularization or amputation for peripheral artery disease, or cardiovascular death. Preplanned secondary outcomes are all-cause mortality, incidence of the metabolic syndrome, incidence of type 2 diabetes, incidence of renal failure, incidence of abnormal bone mineral density and body fat distribution. Patients participating to the cohort will be eligible to be enrolled in four pre-planned sub-studies of cardiovascular imaging, glucose metabolism, immunological and genetic risk profile. Discussion The Canadian HIV and Aging Cohort will provide insights on pathophysiological pathways leading to premature CVD for patients living with HIV. Electronic supplementary material The online version of this article (10.1186/s12879-017-2692-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Madeleine Durand
- Internal Medicine service, Centre de Recherche du CHUM, Montréal, QC, H2J 1T8, Canada.
| | | | - Jean-Guy Baril
- Clinique médicale urbaine du Quartier latin, Montreal, Canada
| | - Sylvie Trottier
- Clinique médicale urbaine du Quartier latin, Montreal, Canada
| | - Benoit Trottier
- Clinique médicale urbaine du Quartier latin, Montreal, Canada
| | | | - Sharon Walmsley
- Division of Infectious Diseases, University Health Network, Toronto, Canada
| | - Brian Conway
- Division of Infectious Diseases, University Health Network, Toronto, Canada
| | - Alexander Wong
- Infectious Diseases Clinic, Regina Qu'Appelle Health Region, Regina, Canada
| | - Jean-Pierre Routy
- Chronic viral infection service and Division of Hematology, McGill University Health Centre, Montreal, Canada
| | - Colin Kovacs
- Maple Leaf Medical HIV Research Collaborative Inc., Toronto, Canada
| | - Paul A MacPherson
- The Ottawa Hospital Research Institute and the University of Ottawa, Ottawa, Canada
| | | | - Samer Mansour
- The Ottawa Hospital Research Institute and the University of Ottawa, Ottawa, Canada
| | - George Thanassoulis
- Preventive and Genomic Cardiology, McGill University Health Center and Research Institute, Montreal, Canada
| | - Michal Abrahamowicz
- Department of Epidemiology and Biostatistics, McGill University, Montreal, Canada
| | - Zhitong Zhu
- Department of Epidemiology and Biostatistics, McGill University, Montreal, Canada
| | - Christos Tsoukas
- McGill University, Immunology service, Montreal General Hospital, Montreal, Canada
| | | | - Nicole Bernard
- Research Institute of the McGill University Health Center, Division of Experimental Medicine, McGill University, Division of Clinical Immunology, McGill University health Center (MUHC), Chronic Viral Illness Service, Montreal, Canada
| | | | | |
Collapse
|
53
|
Differential effects of antiretrovirals on microbial translocation and gut microbiota composition of HIV-infected patients. J Int AIDS Soc 2017; 20:21526. [PMID: 28362071 PMCID: PMC5467634 DOI: 10.7448/ias.20.1.21526] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Increased bacterial translocation and alterations to gut microbiota composition have been described in HIV infection and contribute to immune activation and inflammation. These effects persist despite combined antiretroviral therapy (cART). However, the contribution of different cART combinations has not yet been investigated. The aim of this study was to analyse the long-term effects of different combinations of cART on bacterial translocation and gut microbiota composition in HIV-infected patients. METHODS We carried out a cross-sectional study of 45 HIV-infected patients on cART, classified as nucleoside reverse transcriptase inhibitors (NRTIs)+ protease inhibitors (PIs) (n = 15), NRTIs+ non-nucleoside reverse transcriptase inhibitors (NNRTIs) (n = 22), and NRTIs+ integrase strand transfer inhibitors (INSTIs) (n = 8). Untreated HIV-infected patients (n = 5) and non-infected volunteers (n = 21) were also included. Soluble markers of bacterial translocation and inflammation were measured and gut microbiota composition was analysed using 16S rDNA pyrosequencing (Illumina MiSeq). RESULTS The NRTIs+INSTIs regimen was associated with levels of systemic inflammation that were similar to uninfected controls. The reduction in faecal bacterial diversity induced by HIV infection was also restored by this regimen. HIV infection was more closely related to changes in lower taxonomic units and diversity rather than at the phylum level. The NRTIs+PIs regimen showed the highest reduction in bacterial species, whereas NRTIs+INSTIs induced a minor loss of bacterial species and a significant increase in others. CONCLUSION Our study demonstrated that INSTI-based ART was associated with levels of systemic inflammation and microbial diversity similar to that of uninfected controls. The role of INSTIs other than raltegravir needs to be further investigated. Patients on the NRTIs+PIs regimen presented the highest reduction in bacterial species compared with other antiretrovirals and naive patients. Thus, different cART regimens are associated with diverse profiles in gut microbiota composition. Longitudinal and functional studies are needed to better understand these findings.
Collapse
|
54
|
Impact of intensified antiretroviral therapy during early HIV infection on gut immunology and inflammatory blood biomarkers. AIDS 2017; 31:1529-1534. [PMID: 28463882 DOI: 10.1097/qad.0000000000001515] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Standard antiretroviral therapy (ART) is slow to reverse gut mucosal immune defects that cause persistent inflammation and immune activation. We examined whether intensifying early-administered ART through the addition of maraviroc and raltegravir would accelerate their resolution. DESIGN ART-naïve men with early HIV infection were randomized in a double-blind manner to receive ART (emtricitabine/tenofovir disoproxil fumarate + lopinavir/ritonavir), together with either combined placebo or raltegravir + maraviroc, for 48 weeks. In a predefined substudy, paired blood and sigmoid biopsies were collected at baseline and week 48. Mucosal CD4 T-cell immune subsets (Th1, Th17, and Th22 cells), CD8 T-cell immune activation, and soluble blood markers of inflammation (IL-6, IL-17, macrophage inflammatory protein-1b, soluble CD14, and IL-10) and coagulation (D-dimer) were measured. RESULTS A total of 22 participants were enrolled, a median of 4 months after HIV acquisition. At baseline, there was substantial systemic and mucosal immune activation, and gut CD4 T-cell numbers, Th22 cell numbers, and Th17 cell function were reduced compared with controls. Early ART restored gut Th22 numbers, improved but did not restore overall CD4 numbers, and had no impact on Th17 function. Plasma levels of soluble CD14 and D-dimer normalized, whereas other inflammatory cytokines were reduced but not normalized. ART intensification had no impact on any blood or gut immune parameters. CONCLUSION Early HIV infection causes substantial mucosal and systemic immune activation, and gut CD4 T-cell dysfunction. One year of ART improved but did not normalize most parameters, regardless of intensification with raltegravir and maraviroc, and did not restore mucosal Th17 function.
Collapse
|
55
|
Abstract
Microbiota play a key role in various body functions, as well as in physiological, metabolic, and immunological processes, through different mechanisms such as the regulation of the development and/or functions of different types of immune cells in the intestines. Evidence indicates that alteration in the gut microbiota can influence infectious and non-infectious diseases. Bacteria that reside on the mucosal surface or within the mucus layer interact with the host immune system, thus, a healthy gut microbiota is essential for the development of mucosal immunity. In patients with human immunodeficiency virus (HIV), including those who control their disease with antiretroviral drugs (ART), the gut microbiome is very different than the microbiome of those not infected with HIV. Recent data suggests that, for these patients, dysbiosis may lead to a breakdown in the gut’s immunologic activity, causing systemic bacteria diffusion and inflammation. Since in HIV-infected patients in this state, including those in ART therapy, the treatment of gastrointestinal tract disorders is frustrating, many studies are in progress to investigate the ability of probiotics to modulate epithelial barrier functions, microbiota composition, and microbial translocation. This mini-review analyzed the use of probiotics to prevent and attenuate several gastrointestinal manifestations and to improve gut-associated lymphoid tissue (GALT) immunity in HIV infection.
Collapse
|
56
|
Lafferty MK, Sun L, Christensen-Quick A, Lu W, Garzino-Demo A. Human Beta Defensin 2 Selectively Inhibits HIV-1 in Highly Permissive CCR6⁺CD4⁺ T Cells. Viruses 2017; 9:v9050111. [PMID: 28509877 PMCID: PMC5454423 DOI: 10.3390/v9050111] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/27/2022] Open
Abstract
Chemokine receptor type 6 (CCR6)⁺CD4⁺ T cells are preferentially infected and depleted during HIV disease progression, but are preserved in non-progressors. CCR6 is expressed on a heterogeneous population of memory CD4⁺ T cells that are critical to mucosal immunity. Preferential infection of these cells is associated, in part, with high surface expression of CCR5, CXCR4, and α4β7. In addition, CCR6⁺CD4⁺ T cells harbor elevated levels of integrated viral DNA and high levels of proliferation markers. We have previously shown that the CCR6 ligands MIP-3α and human beta defensins inhibit HIV replication. The inhibition required CCR6 and the induction of APOBEC3G. Here, we further characterize the induction of apolipoprotein B mRNA editing enzyme (APOBEC3G) by human beta defensin 2. Human beta defensin 2 rapidly induces transcriptional induction of APOBEC3G that involves extracellular signal-regulated kinases 1/2 (ERK1/2) activation and the transcription factors NFATc2, NFATc1, and IRF4. We demonstrate that human beta defensin 2 selectively protects primary CCR6⁺CD4⁺ T cells infected with HIV-1. The selective protection of CCR6⁺CD4⁺ T cell subsets may be critical in maintaining mucosal immune function and preventing disease progression.
Collapse
Affiliation(s)
- Mark K Lafferty
- Division of Basic Science, Institute of Human Virology, 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.
| | - Lingling Sun
- Division of Basic Science, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Aaron Christensen-Quick
- Division of Basic Science, Institute of Human Virology, 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.
| | - Wuyuan Lu
- Division of Basic Science, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Biochemistry, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Alfredo Garzino-Demo
- Division of Basic Science, Institute of Human Virology, 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.
- Department of Molecular Medicine, University of Padova, Padova 35121, Italy.
| |
Collapse
|
57
|
Total HIV-1 DNA, a Marker of Viral Reservoir Dynamics with Clinical Implications. Clin Microbiol Rev 2017; 29:859-80. [PMID: 27559075 DOI: 10.1128/cmr.00015-16] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
HIV-1 DNA persists in infected cells despite combined antiretroviral therapy (cART), forming viral reservoirs. Recent trials of strategies targeting latent HIV reservoirs have rekindled hopes of curing HIV infection, and reliable markers are thus needed to evaluate viral reservoirs. Total HIV DNA quantification is simple, standardized, sensitive, and reproducible. Total HIV DNA load influences the course of the infection and is therefore clinically relevant. In particular, it is predictive of progression to AIDS and death, independently of HIV RNA load and the CD4 cell count. Baseline total HIV DNA load is predictive of the response to cART. It declines during cART but remains quantifiable, at a level that reflects both the history of infection (HIV RNA zenith, CD4 cell count nadir) and treatment efficacy (residual viremia, cumulative viremia, immune restoration, immune cell activation). Total HIV DNA load in blood is also predictive of the presence and severity of some HIV-1-associated end-organ disorders. It can be useful to guide individual treatment, notably, therapeutic de-escalation. Although it does not distinguish between replication-competent and -defective latent viruses, the total HIV DNA load in blood, tissues, and cells provides insights into HIV pathogenesis, probably because all viral forms participate in host cell activation and HIV pathogenesis. Total HIV DNA is thus a biomarker of HIV reservoirs, which can be defined as all infected cells and tissues containing all forms of HIV persistence that participate in pathogenesis. This participation may occur through the production of new virions, creating new cycles of infection and disseminating infected cells; maintenance or amplification of reservoirs by homeostatic cell proliferation; and viral transcription and synthesis of viral proteins without new virion production. These proteins can induce immune activation, thus participating in the vicious circle of HIV pathogenesis.
Collapse
|
58
|
Mzingwane ML, Tiemessen CT. Mechanisms of HIV persistence in HIV reservoirs. Rev Med Virol 2017; 27. [PMID: 28128885 DOI: 10.1002/rmv.1924] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/21/2016] [Accepted: 12/23/2016] [Indexed: 01/18/2023]
Abstract
The establishment and maintenance of HIV reservoirs that lead to persistent viremia in patients on antiretroviral drugs remains the greatest challenge of the highly active antiretroviral therapy era. Cellular reservoirs include resting memory CD4+ T lymphocytes, implicated as the major HIV reservoir, having a half-life of approximately 44 months while this is less than 6 hours for HIV in plasma. In some individuals, persistent viremia consists of invariant HIV clones not detected in circulating resting CD4+ T lymphocytes suggesting other possible sources of residual viremia. Some anatomical reservoirs that may harbor such cells include the brain and the central nervous system, the gastrointestinal tract and the gut-associated lymphoid tissue and other lymphoid organs, and the genital tract. The presence of immune cells and other HIV susceptible cells, occurring in differing compositions in anatomical reservoirs, coupled with variable and poor drug penetration that results in suboptimal drug concentrations in some sites, are all likely factors that fuel the continued low-level replication and persistent viremia during treatment. Latently, HIV-infected CD4+ T cells harboring replication-competent virus, HIV cell-to-cell spread, and HIV-infected T cell homeostatic proliferation due to chronic immune activation represent further drivers of this persistent HIV viremia during highly active antiretroviral therapy.
Collapse
Affiliation(s)
- Mayibongwe L Mzingwane
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Pathology, Faculty of Medicine, National University of Science and Technology, Bulawayo, Zimbabwe
| | - Caroline T Tiemessen
- Centre for HIV and Sexually Transmitted Infections, National Institute for Communicable Diseases, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
59
|
Gosselin A, Wiche Salinas TR, Planas D, Wacleche VS, Zhang Y, Fromentin R, Chomont N, Cohen ÉA, Shacklett B, Mehraj V, Ghali MP, Routy JP, Ancuta P. HIV persists in CCR6+CD4+ T cells from colon and blood during antiretroviral therapy. AIDS 2017; 31:35-48. [PMID: 27835617 PMCID: PMC5131694 DOI: 10.1097/qad.0000000000001309] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/07/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The objective of this article is to investigate the contribution of colon and blood CD4 T-cell subsets expressing the chemokine receptor CCR6 to HIV persistence during antiretroviral therapy. DESIGN Matched sigmoid biopsies and blood samples (n = 13) as well as leukapheresis (n = 20) were collected from chronically HIV-infected individuals receiving antiretroviral therapy. Subsets of CD4 T cells with distinct differentiation/polarization profiles were identified using surface markers as follows: memory (TM, CD45RA), central memory (TCM; CD45RACCR7), effector (TEM/TM; CD45RACCR7), Th17 (CCR6CCR4), Th1Th17 (CCR6CXCR3), Th1 (CCR6CXCR3), and Th2 (CCR6CCR4). METHODS We used polychromatic flow cytometry for cell sorting, nested real-time PCR for HIV DNA quantification, ELISA and flow cytometry for HIV p24 quantification. HIV reactivation was induced by TCR triggering in the presence/absence of all-trans retinoic acid. RESULTS Compared with blood, the frequency of CCR6 TM was higher in the colon. In both colon and blood compartments, CCR6 TM were significantly enriched in HIV DNA when compared with their CCR6 counterparts (n = 13). In blood, integrated HIV DNA levels were significantly enriched in CCR6 versus CCR6 TCM of four of five individuals and CCR6 versus CCR6 TEM of three of five individuals. Among blood TCM, Th17 and Th1Th17 contributed the most to the pool of cells harboring integrated HIV DNA despite their reduced frequency compared with Th2, which were infected the least. HIV reactivation was induced by TCR triggering and/or retinoic acid exposure at higher levels in CCR6 versus CCR6 TM, TCM, and TEM. CONCLUSION CCR6 is a marker for colon and blood CD4 T cells enriched for replication-competent HIV DNA. Novel eradication strategies should target HIV persistence in CCR6CD4 T cells from various anatomic sites.
Collapse
Affiliation(s)
| | - Tomas Raul Wiche Salinas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Delphine Planas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Vanessa S. Wacleche
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Yuwei Zhang
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | | | - Nicolas Chomont
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Éric A. Cohen
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
- Institut de Recherche Clinique de Montréal, Montréal, Québec, Canada
| | | | - Vikram Mehraj
- Chronic Viral Illness Service and Research Institute
| | | | - Jean-Pierre Routy
- Chronic Viral Illness Service and Research Institute
- Division of Hematology, McGill University Health Centre, Montreal, Québec, Canada
| | - Petronela Ancuta
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| |
Collapse
|
60
|
Abstract
HIV-1 infection is associated with substantial damage to the gastrointestinal tract resulting in structural impairment of the epithelial barrier and a disruption of intestinal homeostasis. The accompanying translocation of microbial products and potentially microbes themselves from the lumen into systemic circulation has been linked to immune activation, inflammation, and HIV-1 disease progression. The importance of microbial translocation in the setting of HIV-1 infection has led to a recent focus on understanding how the communities of microbes that make up the intestinal microbiome are altered during HIV-1 infection and how they interact with mucosal immune cells to contribute to inflammation. This review details the dysbiotic intestinal communities associated with HIV-1 infection and their potential link to HIV-1 pathogenesis. We detail studies that begin to address the mechanisms driving microbiota-associated immune activation and inflammation and the various treatment strategies aimed at correcting dysbiosis and improving the overall health of HIV-1-infected individuals. Finally, we discuss how this relatively new field of research can advance to provide a more comprehensive understanding of the contribution of the gut microbiome to HIV-1 pathogenesis.
Collapse
|
61
|
Lu X, Li Z, Li Q, Jiao Y, Ji Y, Zhang H, Liu Z, Li W, Wu H. Preferential loss of gut-homing α4β7 CD4 + T cells and their circulating functional subsets in acute HIV-1 infection. Cell Mol Immunol 2016; 13:776-784. [PMID: 26277899 PMCID: PMC5101442 DOI: 10.1038/cmi.2015.60] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 01/10/2023] Open
Abstract
Preferential infection and depletion of gut-homing α4β7 CD4+ T cells in the blood are observed in chronic HIV/SIV infection. The dynamic change in gut-homing α4β7 CD4+ T cells and their functional subsets during the acute stages of HIV-1 infection are less documented. Therefore, we conducted a cohort study to investigate whether acute HIV-1 infection induced abnormalities in gut-homing α4β7 CD4+ T cells and their functional subsets. We examined the frequency, absolute number, and functionality of gut-homing α4β7 CD4+ T cells in 26 acute HIV-1-infected patients compared with 20 healthy individuals. We found that circulating gut-homing α4β7 CD4+ T cells were preferentially depleted during acute HIV-1 infection and were positively correlated with absolute CD4+ T-cell count in blood. Notably, Th17 and Th1 cell subsets of gut-homing CD4+ T cells were also decreased, which resulted in an imbalance of T helper cells (Th1):regulatory T cells (Treg) and Treg:Th17 ratios. Gut-homing Th17 and Th1 cells were also positively correlated with the absolute number of total CD4+ T cells and gut-homing CD4+ T cells. The gut-homing Treg:Th17 ratio was inversely correlated with the CD4+ T-cell count. Taken together, the analyses of our acute HIV-1 cohort demonstrate that gut-homing α4β7 CD4+ T cells and their functional subsets were profoundly depleted during acute HIV-1 infection, which may have resulted in the persistent loss of circulating CD4+ T cells and an imbalance of Th1:Treg and Treg:Th17 ratios and contribute to HIV-1 disease pathogenesis.
Collapse
Affiliation(s)
- Xiaofan Lu
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Zhen Li
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Qunhui Li
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| | - Yanmei Jiao
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Yunxia Ji
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Hongwei Zhang
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| | - Zhuoming Liu
- Case Comprehensive Cancer Center and Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wei Li
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| | - Hao Wu
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| |
Collapse
|
62
|
Melkova Z, Shankaran P, Madlenakova M, Bodor J. Current views on HIV-1 latency, persistence, and cure. Folia Microbiol (Praha) 2016; 62:73-87. [PMID: 27709447 DOI: 10.1007/s12223-016-0474-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/20/2016] [Indexed: 01/01/2023]
Abstract
HIV-1 infection cannot be cured as it persists in latently infected cells that are targeted neither by the immune system nor by available therapeutic approaches. Consequently, a lifelong therapy suppressing only the actively replicating virus is necessary. The latent reservoir has been defined and characterized in various experimental models and in human patients, allowing research and development of approaches targeting individual steps critical for HIV-1 latency establishment, maintenance, and reactivation. However, additional mechanisms and processes driving the remaining low-level HIV-1 replication in the presence of the suppressive therapy still remain to be identified and targeted. Current approaches toward HIV-1 cure involve namely attempts to reactivate and purge HIV latently infected cells (so-called "shock and kill" strategy), as well as approaches involving gene therapy and/or gene editing and stem cell transplantation aiming at generation of cells resistant to HIV-1. This review summarizes current views and concepts underlying different approaches aiming at functional or sterilizing cure of HIV-1 infection.
Collapse
Affiliation(s)
- Zora Melkova
- Department of Immunology and Microbiology, 1st Faculty of Medicine, Charles University, Studnickova 7, 128 00, Prague 2, Czech Republic. .,BIOCEV, Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec, Průmyslová 595, 252 50, Vestec, Czech Republic.
| | - Prakash Shankaran
- Department of Immunology and Microbiology, 1st Faculty of Medicine, Charles University, Studnickova 7, 128 00, Prague 2, Czech Republic
| | - Michaela Madlenakova
- Department of Immunology and Microbiology, 1st Faculty of Medicine, Charles University, Studnickova 7, 128 00, Prague 2, Czech Republic.,BIOCEV, Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec, Průmyslová 595, 252 50, Vestec, Czech Republic
| | - Josef Bodor
- BIOCEV, Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec, Průmyslová 595, 252 50, Vestec, Czech Republic
| |
Collapse
|
63
|
Crothers K, Petrache I, Wongtrakool C, Lee PJ, Schnapp LM, Gharib SA. Widespread activation of immunity and pro-inflammatory programs in peripheral blood leukocytes of HIV-infected patients with impaired lung gas exchange. Physiol Rep 2016; 4:4/8/e12756. [PMID: 27117807 PMCID: PMC4848721 DOI: 10.14814/phy2.12756] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 03/13/2016] [Indexed: 01/17/2023] Open
Abstract
HIV infection is associated with impaired lung gas transfer as indicated by a low diffusing capacity (DLCO), but the mechanisms are not well understood. We hypothesized that HIV-associated gas exchange impairment is indicative of system-wide perturbations that could be reflected by alterations in peripheral blood leukocyte (PBL) gene expression. Forty HIV-infected (HIV(+)) and uninfected (HIV(-)) men with preserved versus low DLCO were enrolled. All subjects were current smokers and those with acute illness, lung diseases other than COPD or asthma were excluded. Total RNA was extracted from PBLs and hybridized to whole-genome microarrays. Gene set enrichment analysis (GSEA) was performed between HIV(+) versus HIV(-) subjects with preserved DLCO and those with low DLCO to identify differentially activated pathways. Using pathway-based analyses, we found that in subjects with preserved DLCO, HIV infection is associated with activation of processes involved in immunity, cell cycle, and apoptosis. Applying a similar analysis to subjects with low DLCO, we identified a much broader repertoire of pro-inflammatory and immune-related pathways in HIV(+) patients relative to HIV(-) subjects, with up-regulation of multiple interleukin pathways, interferon signaling, and toll-like receptor signaling. We confirmed elevated circulating levels of IL-6 in HIV(+) patients with low DLCO relative to the other groups. Our findings reveal that PBLs of subjects with HIV infection and low DLCO are distinguished by widespread enrichment of immuno-inflammatory programs. Activation of these pathways may alter the biology of circulating leukocytes and play a role in the pathogenesis of HIV-associated gas exchange impairment.
Collapse
Affiliation(s)
- Kristina Crothers
- Division of Pulmonary & Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington
| | - Irina Petrache
- Division of Pulmonary & Critical Care Medicine, National Jewish Health, Denver, Colorado
| | - Cherry Wongtrakool
- Pulmonary Section, Department of Veterans Affairs Medical Center, Decatur, Georgia Division of Pulmonary, Allergy, Critical Care, & Sleep Medicine, Emory University, Atlanta, Georgia
| | - Patty J Lee
- Division of Pulmonary & Critical Care Medicine, Yale University, New Haven, Connecticut
| | - Lynn M Schnapp
- Division of Pulmonary & Critical Care Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Sina A Gharib
- Division of Pulmonary & Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington
| |
Collapse
|
64
|
CCR6(-) regulatory T cells blunt the restoration of gut Th17 cells along the CCR6-CCL20 axis in treated HIV-1-infected individuals. Mucosal Immunol 2016; 9:1137-50. [PMID: 26883727 DOI: 10.1038/mi.2016.7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/10/2016] [Indexed: 02/07/2023]
Abstract
The gut CD4(+) T cells, particularly the T helper type 17 (Th17) subset, are not completely restored in most HIV-1-infected individuals despite combined antiretroviral therapy, when initiated at the chronic phase of infection. We show here that the CCR6-CCL20 chemotactic axis is altered, with reduced CCL20 production by small intestine epithelial cells in treated HIV-1-infected individuals. This leads to impaired CCR6(+)CD4(+) T-cell homing, particularly Th17 cells, to the small intestine mucosa. In contrast, the frequency of gut FoxP3(+) T regulatory (Treg) cells, specifically the CCR6(-) subset, was increased. The resulting imbalance in the Th17/CCR6(-) Treg ratio and the associated shift from interleukin (IL)-17 to IL-10 and transforming growth factor-β (TGF-β) blunts CCL20 production by enterocytes, perpetuating a negative feedback for the recruitment of CCR6(+)CD4(+) T cells to the small intestine in treated HIV-1-infected individuals.
Collapse
|
65
|
Sena AAS, Glavan T, Jiang G, Sankaran-Walters S, Grishina I, Dandekar S, Goulart LR. Divergent Annexin A1 expression in periphery and gut is associated with systemic immune activation and impaired gut immune response during SIV infection. Sci Rep 2016; 6:31157. [PMID: 27484833 PMCID: PMC4971494 DOI: 10.1038/srep31157] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 07/15/2016] [Indexed: 01/18/2023] Open
Abstract
HIV-1 disease progression is paradoxically characterized by systemic chronic immune activation and gut mucosal immune dysfunction, which is not fully defined. Annexin A1 (ANXA1), an inflammation modulator, is a potential link between systemic inflammation and gut immune dysfunction during the simian immunodeficiency virus (SIV) infection. Gene expression of ANXA1 and cytokines were assessed in therapy-naïve rhesus macaques during early and chronic stages of SIV infection and compared with SIV-negative controls. ANXA1 expression was suppressed in the gut but systemically increased during early infection. Conversely, ANXA1 expression increased in both compartments during chronic infection. ANXA1 expression in peripheral blood was positively correlated with HLA-DR+CD4+ and CD8+ T-cell frequencies, and negatively associated with the expression of pro-inflammatory cytokines and CCR5. In contrast, the gut mucosa presented an anergic cytokine profile in relation to ANXA1 expression. In vitro stimulations with ANXA1 peptide resulted in decreased inflammatory response in PBMC but increased activation of gut lymphocytes. Our findings suggest that ANXA1 signaling is dysfunctional in SIV infection, and may contribute to chronic inflammation in periphery and with immune dysfunction in the gut mucosa. Thus, ANXA1 signaling may be a novel therapeutic target for the resolution of immune dysfunction in HIV infection.
Collapse
Affiliation(s)
- Angela A S Sena
- Institute of Genetics and Biochemistry, Federal University of Uberlandia, Uberlandia, MG, Brazil.,Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Tiffany Glavan
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Guochun Jiang
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Sumathi Sankaran-Walters
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Irina Grishina
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Satya Dandekar
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Luiz R Goulart
- Institute of Genetics and Biochemistry, Federal University of Uberlandia, Uberlandia, MG, Brazil.,Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
66
|
Deleage C, Schuetz A, Alvord WG, Johnston L, Hao XP, Morcock DR, Rerknimitr R, Fletcher JL, Puttamaswin S, Phanuphak N, Dewar R, McCune JM, Sereti I, Robb M, Kim JH, Schacker TW, Hunt P, Lifson JD, Ananworanich J, Estes JD. Impact of early cART in the gut during acute HIV infection. JCI Insight 2016; 1:e87065. [PMID: 27446990 PMCID: PMC4951101 DOI: 10.1172/jci.insight.87065] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/19/2016] [Indexed: 01/03/2023] Open
Abstract
Early after HIV infection there is substantial depletion of CD4+ T cells in the gastrointestinal (GI) tract lamina propria (LP), with associated epithelial barrier damage, leading to microbial translocation and systemic inflammation and immune activation. In this study, we analyzed these early events in the GI tract in a cohort of Thai acute HIV-infected patients and determined the effect of early combination antiretroviral treatment (cART). HIV-uninfected and chronically and acutely HIV-infected patients at different Fiebig stages (I-V) underwent colonic biopsies and then received cART. Immunohistochemistry and quantitative image analysis were performed on cross-sectional and longitudinal colon biopsy specimens (day 0 to week 96) to measure GI tract damage (infiltration of polymorphonuclear cells), inflammation (M×1, TNF-α), immune activation (Ki-67), and the CD4+ T cell population in the LP. The magnitude of GI tract damage, immune activation, and inflammation was significantly increased, with significantly depleted CD4+ T cells in the LP in all acutely infected groups prior to cART compared with HIV-uninfected control participants. While most patients treated during acute infection resolved GI tract inflammation and immune activation back to baseline levels after 24 weeks of cART, most acutely infected participants did not restore their CD4+ T cells after 96 weeks of cART.
Collapse
Affiliation(s)
- Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Alexandra Schuetz
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - W. Gregory Alvord
- Statistical Consulting, Data Management Services Inc., Frederick, Maryland, USA
| | - Leslie Johnston
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Xing-Pei Hao
- Pathology and Histotechnology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - David R. Morcock
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | | | - James L.K. Fletcher
- SEARCH, Bangkok, Thailand
- Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Suwanna Puttamaswin
- SEARCH, Bangkok, Thailand
- Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Nittaya Phanuphak
- SEARCH, Bangkok, Thailand
- Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Robin Dewar
- Virus Isolation and Serology Laboratory, Applied and Developmental Research Directorate, Science Applications International Corp., Frederick Inc. National Cancer Institute, Frederick Cancer Research and Development Center, Frederick, Maryland, USA
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, UCSF, San Francisco, California, USA
| | - Irini Sereti
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Merlin Robb
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Jerome H. Kim
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- SEARCH, Bangkok, Thailand
- International Vaccine Institute, Seoul, South Korea
| | - Timothy W. Schacker
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Peter Hunt
- Positive Health Program, Department of Medicine, UCSF, San Francisco, California, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Jintanat Ananworanich
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- SEARCH, Bangkok, Thailand
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | | |
Collapse
|
67
|
Depincé-Berger AE, Vergnon-Miszczycha D, Girard A, Frésard A, Botelho-Nevers E, Lambert C, Del Tedesco E, Genin C, Pozzetto B, Lucht F, Roblin X, Bourlet T, Paul S. Major influence of CD4 count at the initiation of cART on viral and immunological reservoir constitution in HIV-1 infected patients. Retrovirology 2016; 13:44. [PMID: 27363286 PMCID: PMC4929778 DOI: 10.1186/s12977-016-0278-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/20/2016] [Indexed: 02/01/2023] Open
Abstract
Background A persistent immune activation is observed in gut during HIV-1 infection, which is not completely reversed by a combined antiretroviral therapy (cART). The impact of the time of cART initiation may highly influence the size of the viral reservoir and the ratio of CD4+/CD8+ T cells in the gut. In this study, we analyzed the characteristics of HIV rectal reservoir of long-term treated patients, regarding their blood CD4+ T cells count at the time of cART initiation. Results Twenty-four consenting men were enrolled: 9 exhibiting a CD4+ T cells count >350/mm3 (“high-level CD4 group”) and 15 < 350/mm3 (“low-level CD4 group”) in blood, at the start of cART. An immunophenotypical analysis of T and B cells subpopulations was performed in blood and rectal biopsies. HIV cell-associated DNA loads and qualitative intra-cellular RNA were determined in both compartments. The ratio of CD4+/CD8+ T cells was significantly decreased in the blood but not in the rectum of the “low-level CD4 group” of patients. The alteration in β7+ CD4+ T cells homing was higher in this group and was correlated to a low ratio of CD4+/CD8+ T cells in blood. An initiation of cART in men exhibiting a low-level CD4 count was also associated with an alteration of B cells maturation. HIV blood and gut DNA reservoirs were significantly lower in the “high-level CD4 group” of men. A high HIV DNA level was associated to a detectable intracellular HIV RNA in rectum. Conclusions An early initiation of cART could significantly preserve gut immunity and limit the viral reservoir constitution. Electronic supplementary material The online version of this article (doi:10.1186/s12977-016-0278-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne-Emmanuelle Depincé-Berger
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Delphine Vergnon-Miszczycha
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Alexandre Girard
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France
| | - Anne Frésard
- Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Elisabeth Botelho-Nevers
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Claude Lambert
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Emilie Del Tedesco
- Service d'Hépato-Gastroentérologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Christian Genin
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Bruno Pozzetto
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service des Agents Infectieux et d'Hygiène, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Frédéric Lucht
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Xavier Roblin
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service d'Hépato-Gastroentérologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Thomas Bourlet
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France. .,Service des Agents Infectieux et d'Hygiène, Centre Hospitalo-Universitaire, Saint-Étienne, France.
| | - Stéphane Paul
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| |
Collapse
|
68
|
Kim CJ, Walmsley SL, Raboud JM, Kovacs C, Coburn B, Rousseau R, Reinhard R, Rosenes R, Kaul R. Can Probiotics Reduce Inflammation and Enhance Gut Immune Health in People Living with HIV: Study Designs for the Probiotic Visbiome for Inflammation and Translocation (PROOV IT) Pilot Trials. HIV CLINICAL TRIALS 2016; 17:147-57. [PMID: 27267710 DOI: 10.1080/15284336.2016.1184827] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Despite substantial improvements in HIV outcomes with combination antiretroviral therapy (cART), morbidity and mortality remain above population norms. The gut mucosal immune system is not completely restored by cART, and the resultant microbial translocation may contribute to chronic inflammation, inadequate CD4 T-cell recovery, and increased rates of serious non-AIDS events. Since the microbial environment surrounding a CD4 T cell may influence its development and function, we hypothesize that probiotics provided during cART might reduce inflammation and improve gut immune health in HIV-positive treatment-naïve individuals (PROOV IT I) and individuals with suboptimal CD4 recovery on cART (PROOV IT II). METHODS These prospective, double-blinded, randomized, placebo-controlled, multicenter pilot studies will assess the impact of the probiotic Visbiome at 900 billion bacteria daily. Forty HIV positive cART-naïve men will be randomized in the PROOV IT I study, coincident with antiretroviral initiation, and be followed for 24 weeks. In PROOV IT II, 36 men on cART, but with a CD4 T-cell count below 350 cells/mm(3) will be followed for 48 weeks. The primary outcome for both studies is the comparison of blood CD8 T-cell immune activation. Secondary analyses will include comparison of blood inflammatory biomarkers, microbial translocation, blood and gut immunology and HIV levels, the bacterial community composition, diet, intestinal permeability, and the safety, adherence and tolerability of the study product. DISCUSSION These studies will evaluate the ability of probiotics as a safe and tolerable therapeutic intervention to reduce systemic immune activation and to accelerate gut immune restoration in people living with HIV.
Collapse
Affiliation(s)
- Connie J Kim
- a Toronto General Research Institute , University Health Network , Toronto , Canada
| | - Sharon L Walmsley
- a Toronto General Research Institute , University Health Network , Toronto , Canada.,c Department of Immunology, Dalla Lana School of Public Health, and Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | - Janet M Raboud
- a Toronto General Research Institute , University Health Network , Toronto , Canada.,c Department of Immunology, Dalla Lana School of Public Health, and Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | | | - Bryan Coburn
- a Toronto General Research Institute , University Health Network , Toronto , Canada.,c Department of Immunology, Dalla Lana School of Public Health, and Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | - Rodney Rousseau
- c Department of Immunology, Dalla Lana School of Public Health, and Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | | | - Ron Rosenes
- e Community Health Advocate and Consultant , Toronto , ON , Canada
| | - Rupert Kaul
- a Toronto General Research Institute , University Health Network , Toronto , Canada.,c Department of Immunology, Dalla Lana School of Public Health, and Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| |
Collapse
|
69
|
Massanella M, Fromentin R, Chomont N. Residual inflammation and viral reservoirs: alliance against an HIV cure. Curr Opin HIV AIDS 2016; 11:234-41. [PMID: 26575148 PMCID: PMC4743501 DOI: 10.1097/coh.0000000000000230] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW HIV persists in cellular and anatomical reservoirs during antiretroviral therapy (ART). Viral persistence is ensured by a variety of mechanisms including ongoing viral replication and proliferation of latently infected cells. In this review, we summarize recent findings establishing a link between the unresolved levels of inflammation observed in virally suppressed individuals on ART and the mechanisms responsible for HIV persistence. RECENT FINDINGS Residual levels of viral replication during ART are associated with persistent low levels of immune activation, suggesting that unresolved inflammation can promote the replenishment of the HIV reservoir in tissues. In addition, the recent findings that the latent HIV reservoir is maintained by continuous proliferation of latently infected cells provide another mechanism by which residual inflammation could contribute to HIV persistence. SUMMARY Residual inflammation during ART is likely to be a critical parameter contributing to HIV persistence. Therefore, reducing inflammation may be an efficient way to interfere with the maintenance of the HIV reservoir in virally suppressed individuals on ART.
Collapse
Affiliation(s)
- Marta Massanella
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| | - Rémi Fromentin
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| | - Nicolas Chomont
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| |
Collapse
|
70
|
Denton PW, Søgaard OS, Tolstrup M. Using animal models to overcome temporal, spatial and combinatorial challenges in HIV persistence research. J Transl Med 2016; 14:44. [PMID: 26861779 PMCID: PMC4746773 DOI: 10.1186/s12967-016-0807-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/29/2016] [Indexed: 12/03/2022] Open
Abstract
Research challenges associated with understanding HIV persistence during antiretroviral therapy can be categorized as temporal, spatial and combinatorial. Temporal research challenges relate to the timing of events during establishment and maintenance of HIV persistence. Spatial research challenges regard the anatomical locations and cell subsets that harbor persistent HIV. Combinatorial research challenges pertain to the order of administration, timing of administration and specific combinations of compounds to be administered during HIV eradication therapy. Overcoming these challenges will improve our understanding of HIV persistence and move the field closer to achieving eradication of persistent HIV. Given that humanized mice and non-human primate HIV models permit rigorous control of experimental conditions, these models have been used extensively as in vivo research platforms for directly addressing these research challenges. The aim of this manuscript is to provide a comprehensive review of these recent translational advances made in animal models of HIV persistence.
Collapse
Affiliation(s)
- Paul W Denton
- Institute of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark. .,Department of Infectious Diseases, Aarhus University Hospital, Skejby, Aarhus, Denmark. .,Aarhus Institute for Advanced Studies, Aarhus University, Aarhus, Denmark.
| | - Ole S Søgaard
- Institute of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark. .,Department of Infectious Diseases, Aarhus University Hospital, Skejby, Aarhus, Denmark.
| | - Martin Tolstrup
- Institute of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark. .,Department of Infectious Diseases, Aarhus University Hospital, Skejby, Aarhus, Denmark.
| |
Collapse
|
71
|
Arnold KB, Burgener A, Birse K, Romas L, Dunphy LJ, Shahabi K, Abou M, Westmacott GR, McCorrister S, Kwatampora J, Nyanga B, Kimani J, Masson L, Liebenberg LJ, Abdool Karim SS, Passmore JAS, Lauffenburger DA, Kaul R, McKinnon LR. Increased levels of inflammatory cytokines in the female reproductive tract are associated with altered expression of proteases, mucosal barrier proteins, and an influx of HIV-susceptible target cells. Mucosal Immunol 2016; 9:194-205. [PMID: 26104913 DOI: 10.1038/mi.2015.51] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/04/2015] [Indexed: 02/04/2023]
Abstract
Elevated inflammatory cytokines (EMCs) at mucosal surfaces have been associated with HIV susceptibility, but the underlying mechanisms remain unclear. We characterized the soluble mucosal proteome associated with elevated cytokine expression in the female reproductive tract. A scoring system was devised based on the elevation (upper quartile) of at least three of seven inflammatory cytokines in cervicovaginal lavage. Using this score, HIV-uninfected Kenyan women were classified as either having EMC (n=28) or not (n=68). Of 455 proteins quantified in proteomic analyses, 53 were associated with EMC (5% false discovery rate threshold). EMCs were associated with proteases, cell motility, and actin cytoskeletal pathways, whereas protease inhibitor, epidermal cell differentiation, and cornified envelope pathways were decreased. Multivariate analysis identified an optimal signature of 16 proteins that distinguished the EMC group with 88% accuracy. Three proteins in this signature were neutrophil-associated proteases that correlated with many cytokines, especially GM-CSF (granulocyte-macrophage colony-stimulating factor), IL-1β (interleukin-1β), MIP-3α (macrophage inflammatory protein-3α), IL-17, and IL-8. Gene set enrichment analyses implicated activated immune cells; we verified experimentally that EMC women had an increased frequency of endocervical CD4(+) T cells. These data reveal strong linkages between mucosal cytokines, barrier function, proteases, and immune cell movement, and propose these as potential mechanisms that increase risk of HIV acquisition.
Collapse
Affiliation(s)
- Kelly B Arnold
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Burgener
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.,Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Kenzie Birse
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Laura Romas
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Laura J Dunphy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Max Abou
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Garrett R Westmacott
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Stuart McCorrister
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Jessie Kwatampora
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Billy Nyanga
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Joshua Kimani
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Lindi Masson
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Lenine J Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,Mailman School of Public Health, Department of Epidemiology, Columbia University, New York, USA
| | - Jo-Ann S Passmore
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,National Health Laboratory Service, South Africa
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya.,University Health Network, University of Toronto, Toronto, Canada
| | - Lyle R McKinnon
- Department of Medicine, University of Toronto, Toronto, Canada.,Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya.,Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| |
Collapse
|
72
|
Cleret-Buhot A, Zhang Y, Planas D, Goulet JP, Monteiro P, Gosselin A, Wacleche VS, Tremblay CL, Jenabian MA, Routy JP, El-Far M, Chomont N, Haddad EK, Sekaly RP, Ancuta P. Identification of novel HIV-1 dependency factors in primary CCR4(+)CCR6(+)Th17 cells via a genome-wide transcriptional approach. Retrovirology 2015; 12:102. [PMID: 26654242 PMCID: PMC4676116 DOI: 10.1186/s12977-015-0226-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/22/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The HIV-1 infection is characterized by profound CD4(+) T cell destruction and a marked Th17 dysfunction at the mucosal level. Viral suppressive antiretroviral therapy restores Th1 but not Th17 cells. Although several key HIV dependency factors (HDF) were identified in the past years via genome-wide siRNA screens in cell lines, molecular determinants of HIV permissiveness in primary Th17 cells remain to be elucidated. RESULTS In an effort to orient Th17-targeted reconstitution strategies, we investigated molecular mechanisms of HIV permissiveness in Th17 cells. Genome-wide transcriptional profiling in memory CD4(+) T-cell subsets enriched in cells exhibiting Th17 (CCR4(+)CCR6(+)), Th1 (CXCR3(+)CCR6(-)), Th2 (CCR4(+)CCR6(-)), and Th1Th17 (CXCR3(+)CCR6(+)) features revealed remarkable transcriptional differences between Th17 and Th1 subsets. The HIV-DNA integration was superior in Th17 versus Th1 upon exposure to both wild-type and VSV-G-pseudotyped HIV; this indicates that post-entry mechanisms contribute to viral replication in Th17. Transcripts significantly enriched in Th17 versus Th1 were previously associated with the regulation of TCR signaling (ZAP-70, Lck, and CD96) and Th17 polarization (RORγt, ARNTL, PTPN13, and RUNX1). A meta-analysis using the NCBI HIV Interaction Database revealed a set of Th17-specific HIV dependency factors (HDFs): PARG, PAK2, KLF2, ITGB7, PTEN, ATG16L1, Alix/AIP1/PDCD6IP, LGALS3, JAK1, TRIM8, MALT1, FOXO3, ARNTL/BMAL1, ABCB1/MDR1, TNFSF13B/BAFF, and CDKN1B. Functional studies demonstrated an increased ability of Th17 versus Th1 cells to respond to TCR triggering in terms of NF-κB nuclear translocation/DNA-binding activity and proliferation. Finally, RNA interference studies identified MAP3K4 and PTPN13 as two novel Th17-specific HDFs. CONCLUSIONS The transcriptional program of Th17 cells includes molecules regulating HIV replication at multiple post-entry steps that may represent potential targets for novel therapies aimed at protecting Th17 cells from infection and subsequent depletion in HIV-infected subjects.
Collapse
Affiliation(s)
- Aurélie Cleret-Buhot
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Yuwei Zhang
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Delphine Planas
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | | | - Patricia Monteiro
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Annie Gosselin
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Vanessa Sue Wacleche
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Cécile L Tremblay
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Mohammad-Ali Jenabian
- Département des sciences biologiques, Université du Québec à Montréal, Montreal, QC, Canada.
| | - Jean-Pierre Routy
- Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada. .,Research Institute, McGill University Health Centre, Montreal, QC, Canada. .,Division of Hematology, McGill University Health Centre, Montreal, QC, Canada.
| | - Mohamed El-Far
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Nicolas Chomont
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| | - Elias K Haddad
- Division of infectious Diseases and HIV Medicine, Drexel University, Philadelphia, PA, USA.
| | | | - Petronela Ancuta
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada. .,CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QUÉBEC, H2X 0A9, Canada.
| |
Collapse
|
73
|
Yang GB. Intestinal tract and acquired immunodeficiency syndrome. Shijie Huaren Xiaohua Zazhi 2015; 23:4304-4316. [DOI: 10.11569/wcjd.v23.i27.4304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The intestinal tract is closely associated with the transmission, disease progression and the prevention and control of acquired immune deficiency syndrome (AIDS). It has been noticed early in AIDS research that a large percent of AIDS patients presented abnormalities in their intestinal tract, such as diarrhea. Now it is known that the intestinal tract has close and complex relationships with AIDS: (1) the intestinal tract is directly involved in the transmission of human immunodeficiency virus-1 (HIV-1); (2) the damage of the intestinal barrier of HIV/AIDS patients directly promotes AIDS disease progression; and (3) most importantly, the intestinal tract is an important target for the treatment and prevention of HIV/AIDS. The author has previously reviewed the progress in understanding the roles of the intestinal tract in HIV-1 infection and the changes of the intestinal tract after HIV-1 infection. In the current review, I discuss the progress in understanding the roles of the damage of the intestinal mucosal immune system in AIDS disease progression, and the potential application value of the restoration of intestinal mucosal immunity in the treatment of AIDS.
Collapse
|
74
|
Analysis of Th17 and Tc17 Frequencies and Antiviral Defenses in Gut-Associated Lymphoid Tissue of Chronic HIV-1 Positive Patients. Mediators Inflamm 2015. [PMID: 26221062 PMCID: PMC4499407 DOI: 10.1155/2015/395484] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complex relationship between both the Th1/Th17 and Tc1/Tc17 axis and innate defences in the intestinal mucosa during HIV-1 infection has not been well characterized. This study examined the frequency, phenotype, and functional status of T cell populations in the gut-associated lymphoid tissue and peripheral blood of virologically suppressed HIV-1-infected patients on therapy, focusing on the Th1, Th17, Tc1, and Tc17 cell subsets. We found a persistent immune cell activation (CD38 and HLADR expression) into the GALT despite the higher levels of Th17 and Tc17 in respect to peripheral blood. An upregulation of type I IFN response in GALT compared to the peripheral blood compartment was also recorded. Furthermore, IFN-α/β levels were negatively related to the frequencies of Th1 naïve cells and Tc1 cell subsets (naïve, central memory, and effector memory) in the GALT. In contrast, no relationships between type I IFN response and Th1 or Tc1 cell subsets in peripheral blood compartment and between IFN-α/β and Th17/Tc17 in both GALT and peripheral blood district were recorded. These data indicate that prolonged antiretroviral treatment improves GALT immune function despite the persistence of immune activation and type I IFN response in chronic HIV-1 positive patients.
Collapse
|
75
|
Aiamkitsumrit B, Sullivan NT, Nonnemacher MR, Pirrone V, Wigdahl B. Human Immunodeficiency Virus Type 1 Cellular Entry and Exit in the T Lymphocytic and Monocytic Compartments: Mechanisms and Target Opportunities During Viral Disease. Adv Virus Res 2015; 93:257-311. [PMID: 26111588 DOI: 10.1016/bs.aivir.2015.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the course of human immunodeficiency virus type 1 infection, a number of cell types throughout the body are infected, with the majority of cells representing CD4+ T cells and cells of the monocyte-macrophage lineage. Both types of cells express, to varying levels, the primary receptor molecule, CD4, as well as one or both of the coreceptors, CXCR4 and CCR5. Viral tropism is determined by both the coreceptor utilized for entry and the cell type infected. Although a single virus may have the capacity to infect both a CD4+ T cell and a cell of the monocyte-macrophage lineage, the mechanisms involved in both the entry of the virus into the cell and the viral egress from the cell during budding and viral release differ depending on the cell type. These host-virus interactions and processes can result in the differential targeting of different cell types by selected viral quasispecies and the overall amount of infectious virus released into the extracellular environment or by direct cell-to-cell spread of viral infectivity. This review covers the major steps of virus entry and egress with emphasis on the parts of the replication process that lead to differences in how the virus enters, replicates, and buds from different cellular compartments, such as CD4+ T cells and cells of the monocyte-macrophage lineage.
Collapse
Affiliation(s)
- Benjamas Aiamkitsumrit
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Neil T Sullivan
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Vanessa Pirrone
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
76
|
Impaired Th17 polarization of phenotypically naive CD4(+) T-cells during chronic HIV-1 infection and potential restoration with early ART. Retrovirology 2015; 12:38. [PMID: 25924895 PMCID: PMC4438463 DOI: 10.1186/s12977-015-0164-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/02/2015] [Indexed: 02/07/2023] Open
Abstract
Background Depletion of mucosal Th17 cells during HIV/SIV infections is a major cause for microbial translocation, chronic immune activation, and disease progression. Mechanisms contributing to Th17 deficit are not fully elucidated. Here we investigated alterations in the Th17 polarization potential of naive-like CD4+ T-cells, depletion of Th17-commited subsets during HIV pathogenesis, and Th17 restoration in response to antiretroviral therapy (ART). Results Peripheral blood CD4+ T-cells expressing a naive-like phenotype (CD45RA+CCR7+) from chronically HIV-infected subjects receiving ART (CI on ART; median CD4 counts 592 cells/μl; viral load: <50 HIV-RNA copies/ml; time since infection: 156 months) compared to uninfected controls (HIV-) were impaired in their survival and Th17 polarization potential in vitro. In HIV- controls, IL-17A-producing cells mainly originated from naive-like T-cells with a regulatory phenotype (nTregs: CD25highCD127−FoxP3+) and from CD25+CD127+FoxP3− cells (DP, double positive). Th17-polarized conventional naive CD4+ T-cells (nT: CD25−CD127+FoxP3−) also produced IL17A, but at lower frequency compared to nTregs and DP. In CI on ART subjects, the frequency/counts of nTreg and DP were significantly diminished compared to HIV- controls, and this paucity was further associated with decreased proportions of memory T-cells producing IL-17A and expressing Th17 markers (CCR6+CD26+CD161+, mTh17). nTregs and DP compared to nT cells harbored superior levels of integrated/non-integrated HIV-DNA in CI on ART subjects, suggesting that permissiveness to integrative/abortive infection contributes to impaired survival and Th17 polarization of lineage-committed cells. A cross-sectional study in CI on ART subjects revealed that nTregs, DP and mTh17 counts were negatively correlated with the time post-infection ART was initiated and positively correlated with nadir CD4 counts. Finally, a longitudinal analysis in a HIV primary infection cohort demonstrated a tendency for increased nTreg, DP, and mTh17 counts with ART initiation during the first year of infection. Conclusions These results support a model in which the paucity of phenotypically naive nTregs and DP cells, caused by integrative/abortive HIV infection and/or other mechanisms, contributes to Th17 deficiency in HIV-infected subjects. Early ART initiation, treatment intensification with integrase inhibitors, and/or other alternative interventions aimed at preserving/restoring the pool of cells prone to acquire Th17 functions may significantly improve mucosal immunity in HIV-infected subjects. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0164-6) contains supplementary material, which is available to authorized users.
Collapse
|
77
|
Attia EF, Akgün KM, Wongtrakool C, Goetz MB, Rodriguez-Barradas MC, Rimland D, Brown ST, Soo Hoo GW, Kim J, Lee PJ, Schnapp LM, Sharafkhaneh A, Justice AC, Crothers K. Increased risk of radiographic emphysema in HIV is associated with elevated soluble CD14 and nadir CD4. Chest 2015; 146:1543-1553. [PMID: 25080158 DOI: 10.1378/chest.14-0543] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The association between HIV and emphysema remains incompletely understood. We sought to determine whether HIV is an independent risk factor for emphysema severity and whether markers of HIV severity and systemic biomarkers of inflammation (IL-6), altered coagulation (D-dimer), and immune activation (soluble CD14) are associated with emphysema. METHODS We performed a cross-sectional analysis of 114 participants with HIV infection and 89 participants without HIV infection in the Examinations of HIV-Associated Lung Emphysema (EXHALE) study. Participants underwent chest CT imaging with blinded semiquantitative interpretation of emphysema severity, distribution, and type. We generated multivariable logistic regression models to determine the risk of HIV for radiographic emphysema, defined as > 10% lung involvement. Similar analyses examined associations of plasma biomarkers, HIV RNA, and recent and nadir CD4 cell counts with emphysema among participants with HIV infection. RESULTS Participants with HIV infection had greater radiographic emphysema severity with increased lower lung zone and diffuse involvement. HIV was associated with significantly increased risk for > 10% emphysema in analyses adjusted for cigarette smoking pack-years (OR, 2.24; 95% CI, 1.12-4.48). In multivariable analyses restricted to participants with HIV infection, nadir CD4 < 200 cells/μL (OR, 2.98; 95% CI, 1.14-7.81), and high soluble CD14 level (upper 25th percentile) (OR, 2.55; 95% CI, 1.04-6.22) were associated with increased risk of > 10% emphysema. IL-6 and D-dimer were not associated with emphysema in HIV. CONCLUSIONS HIV is an independent risk factor for radiographic emphysema. Emphysema severity was significantly greater among participants with HIV infection. Among those with HIV, nadir CD4 < 200 cells/μL and elevated soluble CD14 level were associated with emphysema, highlighting potential mechanisms linking HIV with emphysema.
Collapse
Affiliation(s)
- Engi F Attia
- Department of Medicine, University of Washington, Seattle, WA
| | - Kathleen M Akgün
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, West Haven, CT; Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Cherry Wongtrakool
- Atlanta Veterans Affairs Medical Center, Atlanta, GA; Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Matthew Bidwell Goetz
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA; Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Maria C Rodriguez-Barradas
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX; Department of Medicine, Baylor College of Medicine, Houston, TX
| | - David Rimland
- Atlanta Veterans Affairs Medical Center, Atlanta, GA; Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Sheldon T Brown
- Department of Medicine, James J. Peters Veterans Affairs Medical Center, Bronx, NY; Department of Medicine, Icahn School of Medicine at Mt Sinai, New York, NY
| | - Guy W Soo Hoo
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA; Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Joon Kim
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Patty J Lee
- Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Lynn M Schnapp
- Department of Medicine, University of Washington, Seattle, WA
| | - Amir Sharafkhaneh
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX; Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Amy C Justice
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, West Haven, CT; Department of Medicine, Yale School of Medicine, New Haven, CT
| | | |
Collapse
|
78
|
Phetsouphanh C, Xu Y, Zaunders J. CD4 T Cells Mediate Both Positive and Negative Regulation of the Immune Response to HIV Infection: Complex Role of T Follicular Helper Cells and Regulatory T Cells in Pathogenesis. Front Immunol 2015; 5:681. [PMID: 25610441 PMCID: PMC4285174 DOI: 10.3389/fimmu.2014.00681] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/17/2014] [Indexed: 12/13/2022] Open
Abstract
HIV-1 infection results in chronic activation of cells in lymphoid tissue, including T cells, B-cells, and myeloid lineage cells. The resulting characteristic hyperplasia is an amalgam of proliferating host immune cells in the adaptive response, increased concentrations of innate response mediators due to viral and bacterial products, and homeostatic responses to inflammation. While it is generally thought that CD4 T cells are greatly depleted, in fact, two types of CD4 T cells appear to be increased, namely, regulatory T cells (Tregs) and T follicular helper cells (Tfh). These cells have opposing roles, but may both be important in the pathogenic process. Whether Tregs are failing in their role to limit lymphocyte activation is unclear, but there is no doubt now that Tfh are associated with B-cell hyperplasia and increased germinal center activity. Antiretroviral therapy may reduce the lymphocyte activation, but not completely, and therefore, there is a need for interventions that selectively enhance normal CD4 function without exacerbating Tfh, B-cell, or Treg dysfunction.
Collapse
Affiliation(s)
- Chansavath Phetsouphanh
- Centre for Applied Medical Research, Kirby Institute, St Vincent's Hospital, University of New South Wales , Sydney, NSW , Australia
| | - Yin Xu
- Centre for Applied Medical Research, Kirby Institute, St Vincent's Hospital, University of New South Wales , Sydney, NSW , Australia
| | - John Zaunders
- Centre for Applied Medical Research, Kirby Institute, St Vincent's Hospital, University of New South Wales , Sydney, NSW , Australia
| |
Collapse
|
79
|
Kök A, Hocqueloux L, Hocini H, Carrière M, Lefrou L, Guguin A, Tisserand P, Bonnabau H, Avettand-Fenoel V, Prazuck T, Katsahian S, Gaulard P, Thiébaut R, Lévy Y, Hüe S. Early initiation of combined antiretroviral therapy preserves immune function in the gut of HIV-infected patients. Mucosal Immunol 2015; 8:127-40. [PMID: 24985081 DOI: 10.1038/mi.2014.50] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/15/2014] [Indexed: 02/04/2023]
Abstract
Massive loss of lamina propria CD4(+) T cells, changes in the lymphatic architecture, and altered intestinal epithelial barrier leading to microbial translocation are the common features of HIV-1 infection and are not fully restored under combined antiretroviral therapy (cART). To better understand determinants of gut mucosal restoration, we have performed phenotypic and gene expression analyses of the gut from HIV-infected patients, naive or treated with cART initiated either at the early phase of the primary infection or later during the chronic phase. We found a depletion of T helper type 22 (Th22) and interleukin-17-producing cells in naive patients. These populations, except Th22 cells, were not restored under cART. Regulatory T cells/Th17 ratio was significantly increased in HIV-infected patients and was inversely correlated to the restoration of CD4(+) T cells but not to gut HIV DNA levels. Gene profile analysis of gut mucosal distinguished two groups of patients, which fitted with the timing of cART initiation. In their majority early, but not later treated patients, exhibited conserved intestinal lymphoid structure, epithelial barrier integrity and dendritic cell maturation pathways. Our data demonstrate that early initiation of cART helps to preserve and/or restore lymphoid gut mucosal homeostasis and provide a rationale for initiating cART during the acute phase of HIV infection.
Collapse
Affiliation(s)
- A Kök
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [3] Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - L Hocqueloux
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans, France
| | - H Hocini
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | | | - L Lefrou
- Service d'Hépato-Gastro-Entérologie, CHR d'Orléans-La Source, Orléans, France
| | - A Guguin
- 1] INSERM U955, Team 16, Créteil, France [2] Plateforme de Cytométrie en flux, IMRB, UFR de Médecine, Créteil, France
| | - P Tisserand
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - H Bonnabau
- 1] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [2] INSERM U897 - INRIA SISTM - Univ. Bordeaux Segalen ISPED, Bordeaux, France
| | - V Avettand-Fenoel
- AP-HP, CHU Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - T Prazuck
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans, France
| | - S Katsahian
- Assistance publique -Hôpitaux de Paris (AP-HP)-Hôpital Henri Mondor-Université Paris-Est Créteil, Val-de Marne, France
| | - P Gaulard
- 1] Université Paris Est Créteil, Faculté de Médecine, Créteil, France [2] INSERM U955, Team 9, Créteil, France [3] Département de Pathologie, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| | - R Thiébaut
- 1] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [2] INSERM U897 - INRIA SISTM - Univ. Bordeaux Segalen ISPED, Bordeaux, France
| | - Y Lévy
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [3] Université Paris Est Créteil, Faculté de Médecine, Créteil, France [4] Service d'Immunologie Clinique, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| | - S Hüe
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [3] Université Paris Est Créteil, Faculté de Médecine, Créteil, France [4] Service d'Immunologie Biologique, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
80
|
Epple HJ, Schneider T, Zeitz M. Microbial Translocation and the Effects of HIV/SIV Infection on Mucosal Barrier Function. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00077-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
81
|
Schuetz A, Deleage C, Sereti I, Rerknimitr R, Phanuphak N, Phuang-Ngern Y, Estes JD, Sandler NG, Sukhumvittaya S, Marovich M, Jongrakthaitae S, Akapirat S, Fletscher JLK, Kroon E, Dewar R, Trichavaroj R, Chomchey N, Douek DC, O′Connell RJ, Ngauy V, Robb ML, Phanuphak P, Michael NL, Excler JL, Kim JH, de Souza MS, Ananworanich J. Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV-related immune activation. PLoS Pathog 2014; 10:e1004543. [PMID: 25503054 PMCID: PMC4263756 DOI: 10.1371/journal.ppat.1004543] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023] Open
Abstract
Mucosal Th17 cells play an important role in maintaining gut epithelium integrity and thus prevent microbial translocation. Chronic HIV infection is characterized by mucosal Th17 cell depletion, microbial translocation and subsequent immune-activation, which remain elevated despite antiretroviral therapy (ART) correlating with increased mortality. However, when Th17 depletion occurs following HIV infection is unknown. We analyzed mucosal Th17 cells in 42 acute HIV infection (AHI) subjects (Fiebig (F) stage I-V) with a median duration of infection of 16 days and the short-term impact of early initiation of ART. Th17 cells were defined as IL-17+ CD4+ T cells and their function was assessed by the co-expression of IL-22, IL-2 and IFNγ. While intact during FI/II, depletion of mucosal Th17 cell numbers and function was observed during FIII correlating with local and systemic markers of immune-activation. ART initiated at FI/II prevented loss of Th17 cell numbers and function, while initiation at FIII restored Th17 cell numbers but not their polyfunctionality. Furthermore, early initiation of ART in FI/II fully reversed the initially observed mucosal and systemic immune-activation. In contrast, patients treated later during AHI maintained elevated mucosal and systemic CD8+ T-cell activation post initiation of ART. These data support a loss of Th17 cells at early stages of acute HIV infection, and highlight that studies of ART initiation during early AHI should be further explored to assess the underlying mechanism of mucosal Th17 function preservation.
Collapse
Affiliation(s)
- Alexandra Schuetz
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory of Cancer Research, Frederick, Maryland, United States of America
| | - Irini Sereti
- Clinical and Molecular Retrovirology Section/Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rungsun Rerknimitr
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nittaya Phanuphak
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Yuwadee Phuang-Ngern
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory of Cancer Research, Frederick, Maryland, United States of America
| | - Netanya G. Sandler
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Suchada Sukhumvittaya
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Mary Marovich
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Surat Jongrakthaitae
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Siriwat Akapirat
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | | | - Eugene Kroon
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
- SEARCH, Bangkok, Thailand
| | - Robin Dewar
- Virus Isolation and Serology Laboratory Applied and Developmental Research Directorate Science Applications International Corporation, Frederick, Inc. National Cancer Institute, Frederick Cancer Research and Development Center, Frederick, Maryland, United States of America
| | - Rapee Trichavaroj
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Nitiya Chomchey
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert J. O′Connell
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Viseth Ngauy
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
| | - Merlin L. Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Praphan Phanuphak
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Nelson L. Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jean-Louis Excler
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jerome H. Kim
- SEARCH, Bangkok, Thailand
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Mark S. de Souza
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences – United States Component, Bangkok, Thailand
- SEARCH, Bangkok, Thailand
| | - Jintanat Ananworanich
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- SEARCH, Bangkok, Thailand
- The Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | | |
Collapse
|
82
|
Page EE, Greathead L, Metcalf R, Clark SA, Hart M, Fuchs D, Pantelidis P, Gotch F, Pozniak A, Nelson M, Boasso A, Gazzard B, Kelleher P. Loss of Th22 cells is associated with increased immune activation and IDO-1 activity in HIV-1 infection. J Acquir Immune Defic Syndr 2014; 67:227-35. [PMID: 25314246 DOI: 10.1097/qai.0000000000000294] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Immune activation plays a key role in the immunopathogenesis of HIV-1 infection. Microbial translocation, secondary to loss of epithelial integrity and mucosal immune deficiency, is believed to contribute to systemic immune activation. Interleukin 22 maintains intestinal epithelial barrier integrity and stimulates the secretion of antimicrobial peptides that limit bacterial dissemination and intestinal inflammation. Interleukin 22 is secreted by CD4 T-helper (Th)22 cells independently of interleukin 17A and interferon γ. Th22 cells are characterized by the expression of chemokine receptors (CCR)4, CCR6, and CCR10. METHODS We analyzed the frequency of Th22, Th17, Th1, and CD4 T regulatory (Treg) cells, markers of immune activation (expression of CD38 on CD8 T cells, neopterin, soluble CD14), microbial translocation (lipopolysaccharide-binding protein and 16s ribosomal DNA), and indoleamine 2,3-dioxygenase 1 activity in peripheral blood of antiretroviral therapy (ART)-experienced and ART-naive HIV-1-infected patients and healthy controls. RESULTS We showed a significant reduction in the frequency of Th22 cells in HIV ART-naive patients compared with the healthy controls and HIV ART-experienced patients. We observed a shift away from Th22 and Th17 to Treg cells, which was partially reversed by effective ART. Markers of immune activation negatively correlated with Th22 and Th17 proportions, and with Th22:Treg and Th17:Treg ratios in ART-naive patients. Increased indoleamine 2,3-dioxygenase 1 activity negatively correlated with Th22:Treg and Th17:Treg ratios in the ART-naive group. CONCLUSIONS Loss of Th22 cells and disruption in the balance of Th22 and Treg cells may contribute toward systemic immune activation and mucosal immune deficiency during HIV-1 infection.
Collapse
Affiliation(s)
- Emma E Page
- *Department of Medicine, Imperial College, London, United Kingdom; †Division of Biological Chemistry, Biocenter, Medical University, Innsbruk, Austria; ‡Department of Infection and Immunity, Imperial College NHS Trust, London, United Kingdom; and §Department of HIV and Sexual Health, Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
d'Ettorre G, Baroncelli S, Micci L, Ceccarelli G, Andreotti M, Sharma P, Fanello G, Fiocca F, Cavallari EN, Giustini N, Mallano A, Galluzzo CM, Vella S, Mastroianni CM, Silvestri G, Paiardini M, Vullo V. Reconstitution of intestinal CD4 and Th17 T cells in antiretroviral therapy suppressed HIV-infected subjects: implication for residual immune activation from the results of a clinical trial. PLoS One 2014; 9:e109791. [PMID: 25340778 PMCID: PMC4207675 DOI: 10.1371/journal.pone.0109791] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 09/04/2014] [Indexed: 12/04/2022] Open
Abstract
Introduction During HIV infection the severe depletion of intestinal CD4+ T-cells is associated with microbial translocation, systemic immune activation, and disease progression. This study examined intestinal and peripheral CD4+ T-cell subsets reconstitution under combined antiretroviral therapy (cART), and systemic immune activation markers. Methods This longitudinal single-arm pilot study evaluates CD4+ T cells, including Th1 and Th17, in gut and blood and soluble markers for inflammation in HIV-infected individuals before (M0) and after eight (M8) months of cART. From January 2010 to December 2011, 10 HIV-1 naïve patients were screened and 9 enrolled. Blood and gut CD4+ T-cells subsets and cellular immune activation were determined by flow-cytometry and plasma soluble CD14 by ELISA. CD4+ Th17 cells were detected in gut biopsies by immunohistochemistry. Microbial translocation was measured by limulus-amebocyte-lysate assay to detect bacterial lipopolysaccharide (LPS) and PCR Real Time to detect plasma bacterial 16S rDNA. Results Eight months of cART increased intestinal CD4+ and Th17 cells and reduced levels of T-cell activation and proliferation. The magnitude of intestinal CD4+ T-cell reconstitution correlated with the reduction of plasma LPS. Importantly, the magnitude of Th17 cells reconstitution correlated directly with blood CD4+ T-cell recovery. Conclusion Short-term antiretroviral therapy resulted in a significant increase in the levels of total and Th17 CD4+ T-cells in the gut mucosa and in decline of T-cell activation. The observation that pre-treatment levels of CD4+ and of CD8+ T-cell activation are predictors of the magnitude of Th17 cell reconstitution following cART provides further rationale for an early initiation of cART in HIV-infected individuals. Trial Registration ClinicalTrials.gov NCT02097381
Collapse
Affiliation(s)
- Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Istituto Pasteur – Fondazione Cenci-Bolognetti, University of Rome “Sapienza”, Rome, Italy
- * E-mail:
| | - Silvia Baroncelli
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Luca Micci
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Istituto Pasteur – Fondazione Cenci-Bolognetti, University of Rome “Sapienza”, Rome, Italy
| | - Mauro Andreotti
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Prachi Sharma
- Division of Pathology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Gianfranco Fanello
- Department of Emergency Surgery- Emergency Endoscopic Unit, Policlinico Umberto I, University of Rome “Sapienza”, Rome, Italy
| | - Fausto Fiocca
- Department of Emergency Surgery- Emergency Endoscopic Unit, Policlinico Umberto I, University of Rome “Sapienza”, Rome, Italy
| | - Eugenio Nelson Cavallari
- Department of Public Health and Infectious Diseases, Istituto Pasteur – Fondazione Cenci-Bolognetti, University of Rome “Sapienza”, Rome, Italy
| | - Noemi Giustini
- Department of Public Health and Infectious Diseases, Istituto Pasteur – Fondazione Cenci-Bolognetti, University of Rome “Sapienza”, Rome, Italy
| | - Alessandra Mallano
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Clementina M. Galluzzo
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Vella
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Claudio M. Mastroianni
- Infectious Disease Unit, Fondazione Eleonora Lorillard Spencer Cenci, Sapienza University, Latina, Italy
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Vincenzo Vullo
- Department of Public Health and Infectious Diseases, Istituto Pasteur – Fondazione Cenci-Bolognetti, University of Rome “Sapienza”, Rome, Italy
| |
Collapse
|
84
|
Mucosal immunity in the female genital tract, HIV/AIDS. BIOMED RESEARCH INTERNATIONAL 2014; 2014:350195. [PMID: 25313360 PMCID: PMC4181941 DOI: 10.1155/2014/350195] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/27/2014] [Accepted: 08/28/2014] [Indexed: 01/15/2023]
Abstract
Mucosal immunity consists of innate and adaptive immune responses which can be influenced by systemic immunity. Despite having been the subject of intensive studies, it is not fully elucidated what exactly occurs after HIV contact with the female genital tract mucosa. The sexual route is the main route of HIV transmission, with an increased risk of infection in women compared to men. Several characteristics of the female genital tract make it suitable for inoculation, establishment of infection, and systemic spread of the virus, which causes local changes that may favor the development of infections by other pathogens, often called sexually transmitted diseases (STDs). The relationship of these STDs with HIV infection has been widely studied. Here we review the characteristics of mucosal immunity of the female genital tract, its alterations due to HIV/AIDS, and the characteristics of coinfections between HIV/AIDS and the most prevalent STDs.
Collapse
|
85
|
Zhang WJ, Wang Y, Yu K, Duan JZ, Yao WR, Wang Y, Yang RG, Yang GB. Associated changes in the transcription levels of IL-17A and tight junction-associated genes in the duodenal mucosa of rhesus macaques repeatedly exposed to simian/human immunodeficiency virus. Exp Mol Pathol 2014; 97:225-33. [PMID: 25034324 DOI: 10.1016/j.yexmp.2014.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/12/2014] [Indexed: 11/24/2022]
Abstract
BACKGROUND Mucosal barrier dysfunction might play a key role in HIV/AIDS, yet the early effects of HIV-1 on intestinal mucosal barrier, especially tight junctions (TJ) have not been well addressed. AIMS To investigate the effects of acute HIV-1 infection on the expression of intestinal IL-17A and TJ-associated genes using an NHP-AIDS model. METHODS TaqMan probe real-time RT-PCR methods were established and claudin-1, claudin-3, occludin and zonula occluden-1 (ZO-1) mRNA levels in the duodenal biopsies of rhesus macaques collected before and after rectal exposures to SHIV-SF162P4 were examined and compared with that of IL-17A, IL-6, TGF-β, RORγt, T-bet, Foxp-3 and GATA-3. RESULTS The mRNA levels of TJ-associated genes were statistically significantly reduced soon after viral exposures and the mRNA levels of claudin-1, occludin and ZO-1 in viral positive tissues (from Group I) were lower than that in viral negative tissues (from Group II) after viral exposure. IL-17A mRNA levels were also decreased and positively correlated with the mRNA levels of the TJ-associated genes after viral exposure or infection, although the levels of IL-6, TGF-β and RORγt mRNA showed no statistical difference. The levels of GATA-3 mRNA in tissues collected before viral exposure were statistically different between Group I and Group II animals. The balance between T-bet and GATA-3 mRNA levels in Group II was markedly altered and statistically significantly different from that in Group I. CONCLUSIONS Acute SHIV, and by extension HIV infection could affect the expression of TJ-associated genes, probably through IL-17A and other immune alterations.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing 102206, PR China
| | - Yue Wang
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing 102206, PR China
| | - Kai Yu
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing 102206, PR China
| | - Jia-Zhong Duan
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing 102206, PR China
| | - Wen-Rong Yao
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing 102206, PR China
| | - Yan Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, PR China
| | - Rong-Ge Yang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, PR China
| | - Gui-Bo Yang
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing 102206, PR China.
| |
Collapse
|
86
|
Prodger JL, Hirbod T, Gray R, Kigozi G, Nalugoda F, Galiwango R, Reynolds SJ, Huibner S, Wawer MJ, Serwadda D, Kaul R. HIV Infection in Uncircumcised Men Is Associated With Altered CD8 T-cell Function But Normal CD4 T-cell Numbers in the Foreskin. J Infect Dis 2014; 209:1185-94. [PMID: 24277744 PMCID: PMC3969543 DOI: 10.1093/infdis/jit644] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/29/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-infected (HIV+) men are more susceptible to sexually transmitted infections, and may be superinfected by HIV. We hypothesized that HIV induces immune alterations in the foreskin that may impact the subsequent acquisition/clearance of genital coinfections. METHODS Foreskin tissue and blood were obtained from 70 HIV-uninfected and 20 HIV+ men undergoing circumcision. T cells were characterized by flow cytometry, immunohistochemistry, and polymerase chain reaction. RESULTS There was substantial influx of CD8 T-cells into the foreskins of HIV+ men (108.8 vs 23.1 cells/mm(2); P < .001); but foreskin CD4 T-cell density was unchanged (43.0 vs 33.7/mm(2); P = .67), despite substantial blood depletion (409.0 vs 877.8 cells/µL; P < .001). While frequencies of foreskin C-C chemokine receptor type 5(+) (CCR5(+)) T cells, T regulatory cells, and T-helper 17 cells were unaltered in HIV+ men, CD8 T-cell production of tumor necrosis factor α (TNFα) was decreased. HIV-specific CD8 T cells were present in the foreskins of HIV+ men, although their frequency and function was reduced compared to the blood. CONCLUSIONS Foreskin CD4 T-cell density and CCR5 expression were not reduced during HIV infection, perhaps explaining susceptibility to HIV superinfection. Foreskin CD8 T-cell density was increased, but decreased production of TNFα may enhance susceptibility to genital coinfections in HIV+ men.
Collapse
Affiliation(s)
- Jessica L Prodger
- Clinical Science Division, Department of Medicine, University of Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Petrova MI, van den Broek M, Balzarini J, Vanderleyden J, Lebeer S. Vaginal microbiota and its role in HIV transmission and infection. FEMS Microbiol Rev 2014; 37:762-92. [PMID: 23789590 DOI: 10.1111/1574-6976.12029] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 01/21/2023] Open
Abstract
The urogenital tract appears to be the only niche of the human body that shows clear differences in microbiota between men and women. The female reproductive tract has special features in terms of immunological organization, an epithelial barrier, microbiota, and influence by sex hormones such as estrogen. While the upper genital tract is regarded as free of microorganisms, the vagina is colonized by bacteria dominated by Lactobacillus species, although their numbers vary considerably during life. Bacterial vaginosis is a common pathology characterized by dysbiosis, which increases the susceptibility for HIV infection and transmission. On the other hand, HIV infections are often characterized by a disturbed vaginal microbiota. The endogenous vaginal microbiota may protect against HIV by direct production of antiviral compounds, through blocking of adhesion and transmission by ligands such as lectins, and/or by stimulation of immune responses. The potential role of probiotics in the prevention of HIV infections and associated symptoms, by introducing them to the vaginal and gastrointestinal tract (GIT), is also discussed. Of note, the GIT is a site of considerable HIV replication and CD4(+) T-cell destruction, resulting in both local and systemic inflammation. Finally, genetically engineered lactobacilli show promise as new microbicidal agents against HIV.
Collapse
Affiliation(s)
- Mariya I Petrova
- KU Leuven, Centre of Microbial and Plant Genetics, Leuven, Belgium
| | | | | | | | | |
Collapse
|
88
|
Klatt NR, Chomont N, Douek DC, Deeks SG. Immune activation and HIV persistence: implications for curative approaches to HIV infection. Immunol Rev 2014; 254:326-42. [PMID: 23772629 DOI: 10.1111/imr.12065] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite complete or near-complete suppression of human immunodeficiency virus (HIV) replication with combination antiretroviral therapy, both HIV and chronic inflammation/immune dysfunction persist indefinitely. Untangling the association between the virus and the host immune environment during therapy might lead to novel interventions aimed at either curing the infection or preventing the development of inflammation-associated end-organ disease. Chronic inflammation and immune dysfunction might lead to HIV persistence by causing virus production, generating new target cells, enabling infecting of activated and resting target cells, altering the migration patterns of susceptible target cells, increasing the proliferation of infected cells, and preventing normal HIV-specific clearance mechanisms from function. Chronic HIV production or replication might contribute to persistent inflammation and immune dysfunction. The rapidly evolving data on these issues strongly suggest that a vicious cycle might exist in which HIV persistence causes inflammation that in turn contributes to HIV persistence.
Collapse
Affiliation(s)
- Nichole R Klatt
- Department of Pharmaceutics, Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
89
|
Microbial translocation in HIV infection is associated with dyslipidemia, insulin resistance, and risk of myocardial infarction. J Acquir Immune Defic Syndr 2014; 64:425-33. [PMID: 23797689 DOI: 10.1097/qai.0b013e31829f919d] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Microbial translocation has been suggested to be a driver of immune activation and inflammation. It is hypothesized that microbial translocation may be related to dyslipidemia, insulin resistance, and the risk of coronary heart disease in HIV-infected individuals. DESIGN Cross-sectional study of 60 HIV-infected patients on combination antiretroviral therapy with viral suppression >2 years and 31 healthy age-matched controls. METHODS Lipopolysaccharide (LPS) was analyzed by limulus amebocyte lysate colorimetric assay. Lipids, including cholesterol, low-density lipoprotein (LDL), and triglycerides, were measured. Glucose metabolism was determined using an oral glucose tolerance test. Body composition was determined using whole-body dual-energy x-ray absorptiometry scans and magnetic resonance imaging. The Framingham risk score was used to assess risk of cardiovascular disease and myocardial infarction. RESULTS HIV-infected patients had higher level of LPS compared with controls (64 pg/mL vs. 50 pg/mL, P = 0.002). Likewise, HIV-infected patients had higher triglycerides, LDL, and fasting insulin as well as evidence of lower insulin sensitivity compared with controls. Among HIV-infected patients, high LPS was associated with a higher level of triglycerides and LDL and with lower insulin sensitivity. Importantly, among HIV-infected patients, high LPS was associated with a higher Framingham risk score. CONCLUSIONS HIV-infected patients with suppressed viral replication had increased level of microbial translocation as measured by LPS. LPS was associated with cardiometabolic risk factors and increased Framingham risk score. Hence, the gastrointestinal mucosal barrier may be a potential therapeutic target to prevent dyslipidemia and future cardiovascular complications in HIV infection.
Collapse
|
90
|
Aiamkitsumrit B, Dampier W, Antell G, Rivera N, Martin-Garcia J, Pirrone V, Nonnemacher MR, Wigdahl B. Bioinformatic analysis of HIV-1 entry and pathogenesis. Curr HIV Res 2014; 12:132-61. [PMID: 24862329 PMCID: PMC4382797 DOI: 10.2174/1570162x12666140526121746] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 03/18/2014] [Accepted: 05/06/2014] [Indexed: 02/07/2023]
Abstract
The evolution of human immunodeficiency virus type 1 (HIV-1) with respect to co-receptor utilization has been shown to be relevant to HIV-1 pathogenesis and disease. The CCR5-utilizing (R5) virus has been shown to be important in the very early stages of transmission and highly prevalent during asymptomatic infection and chronic disease. In addition, the R5 virus has been proposed to be involved in neuroinvasion and central nervous system (CNS) disease. In contrast, the CXCR4-utilizing (X4) virus is more prevalent during the course of disease progression and concurrent with the loss of CD4(+) T cells. The dual-tropic virus is able to utilize both co-receptors (CXCR4 and CCR5) and has been thought to represent an intermediate transitional virus that possesses properties of both X4 and R5 viruses that can be encountered at many stages of disease. The use of computational tools and bioinformatic approaches in the prediction of HIV-1 co-receptor usage has been growing in importance with respect to understanding HIV-1 pathogenesis and disease, developing diagnostic tools, and improving the efficacy of therapeutic strategies focused on blocking viral entry. Current strategies have enhanced the sensitivity, specificity, and reproducibility relative to the prediction of co-receptor use; however, these technologies need to be improved with respect to their efficient and accurate use across the HIV-1 subtypes. The most effective approach may center on the combined use of different algorithms involving sequences within and outside of the env-V3 loop. This review focuses on the HIV-1 entry process and on co-receptor utilization, including bioinformatic tools utilized in the prediction of co-receptor usage. It also provides novel preliminary analyses for enabling identification of linkages between amino acids in V3 with other components of the HIV-1 genome and demonstrates that these linkages are different between X4 and R5 viruses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA 19102.
| |
Collapse
|
91
|
Reeves RK, Bosinger SE. Innate Immunity in Simian Immunodeficiency Virus Infection. NATURAL HOSTS OF SIV 2014. [PMCID: PMC7149674 DOI: 10.1016/b978-0-12-404734-1.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The past decade has seen the emergence of innate immunity as a mature field. The study of innate immunity has had a significant impact on the concepts of HIV immunity, pathogenesis, and vaccines. In this chapter, basic concepts of innate immunity at the anatomical, cellular, and molecular levels will be introduced from the perspective of their interplay with HIV and simian immunodeficiency virus (SIV). An emphasis will be placed on studies using SIV/non-human primate (NHP) models that shape current models of HIV pathogenesis. Finally, studies modulating the innate system in vivo in NHPs will be discussed.
Collapse
|
92
|
Imbalances of gut-homing CD4+ T-cell subsets in HIV-1-infected Chinese patients. J Acquir Immune Defic Syndr 2013; 64:25-31. [PMID: 23572009 DOI: 10.1097/qai.0b013e318293a114] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Full reconstitution of CD4 T cells in both peripheral blood and mucosal tissues is a desirable goal of treating AIDS patients. To date, few studies have investigated the potential role of gut-homing CD4 T-cell subsets as biomarkers in assisting Asian populations infected with HIV-1. METHODS A large cross-sectional study was conducted among Chinese patients with focus on the frequency, absolute number, and ratio of gut-homing Th1, Th17, and Treg subsets in 3 groups of age- and gender-matched study subjects: healthy donors, untreated AIDS patients, and antiretroviral therapy (ART)-treated patients with sustained undetectable viral load. RESULTS HIV-1 chronic infection resulted in positively correlated loss of total and gut-homing CD4 T cells (P < 0.001) among patients compared with healthy controls. Profiles of T-cell subsets, however, were different between total and gut-homing CD4 T cells in terms of frequency and absolute number. ART partially restored the frequencies of gut-homing Th1, Th17, and Treg cells but the lost number of gut-homing Th17 cells was found not easily reversible. These changes together with an increased frequency of gut-homing CD4 Treg cells led to dual imbalances of gut-homing Th1/Treg and Th17/Treg ratios, which were negatively correlated with viral load (P = 0.014 and P < 0.001) and hardly restored even by prolonged ART. CONCLUSIONS Our findings provide new insights into the investigation of gut-homing Th1/Treg and Th17/Treg imbalances in AIDS patients, which may have potential implications on the reconstitution of mucosal CD4 T cells.
Collapse
|
93
|
Lederman MM, Funderburg NT, Sekaly RP, Klatt NR, Hunt PW. Residual immune dysregulation syndrome in treated HIV infection. Adv Immunol 2013; 119:51-83. [PMID: 23886064 DOI: 10.1016/b978-0-12-407707-2.00002-3] [Citation(s) in RCA: 259] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antiretroviral therapy has revolutionized the course of HIV infection, improving immune function and decreasing dramatically the mortality and morbidity due to the opportunistic complications of the disease. Nonetheless, even with sustained suppression of HIV replication, many HIV-infected persons experience a syndrome characterized by increased T cell activation and evidence of heightened inflammation and coagulation. This residual immune dysregulation syndrome or RIDS is more common in persons who fail to increase circulating CD4+ T cells to normal levels and in several epidemiologic studies it has been associated with increased morbidity and mortality. These morbid and fatal events are not the typical opportunistic infections and malignancies seen in the early AIDS era but rather comprise a spectrum of cardiovascular events, liver disease, metabolic disorders, kidney disease, bone disease, and a spectrum of malignant complications distinguishable from the opportunistic malignancies that characterized the earlier days of the AIDS epidemic. While immune activation, inflammation, and coagulopathy are characteristic of untreated HIV infection and improve with drug-induced control of HIV replication, the drivers of RIDS in treated HIV infection are incompletely understood. And while inflammation, immune activation, and coagulopathy are more common in treated persons who fail to restore circulating CD4+ T cells, it is not entirely clear how these two phenomena are linked.
Collapse
Affiliation(s)
- Michael M Lederman
- Division of Infectious Diseases and Center for AIDS Research, Case Western Reserve University and University Hospitals/Case Medical Center, Cleveland, Ohio, USA.
| | | | | | | | | |
Collapse
|
94
|
Zaffiri L, d'Ettorre G, Ceccarelli G, Andreotti M, Marcellini S, Giustini N, Ascoli-Bartoli T, Mastroianni CM, Vella S, Vullo V. Recovery of interleukin-17 production from interleukin-15-stimulated CD4+ mononuclear cells in HIV-1-infected patients with sustained viral suppression. J Interferon Cytokine Res 2013; 34:35-40. [PMID: 24102576 DOI: 10.1089/jir.2012.0011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Interleukin-17 (IL-17) is a pro-inflammatory cytokine that is mainly produced by CD4+ T cells. The role of Th17 during the human immunodeficiency virus (HIV)-1 infection is still unclear, but HIV-1 infection can cause a preferential depletion of Th17 cells. It has been shown that IL-15 elicits IL-17 production from human peripheral blood mononuclear cells. We studied the effect of IL-15 stimulation in vitro on IL-17 production from CD4+ mononuclear cells of HIV-infected patients. We observed that IL-15 triggers, in a dose-dependent manner, IL-17 secretion. This effect was blocked by anti-IL-15 monoclonal antibody (P=0.01). Interestingly, IL-17 production was significantly lower in patients with detectable plasma viremia when compared with successfully treated HIV-infected patients (P=0.02) and healthy controls, respectively (P<0.001). We also noticed a significant difference in IL-17 production between naïve HIV-infected patients and patients with virological failure on combined antiretroviral therapy (cART) (P=0.02). Our results suggest that IL-15 can induce IL-17 production from peripheral CD4+ mononuclear cells of HIV-infected patients. Persistent HIV plasma viremia could cause a severe perturbation of IL-17 production from CD4+ mononuclear cells. IL-17 production in HIV-infected patients could be recovered through a sustained suppression of the viral replication in the peripheral blood through cART.
Collapse
Affiliation(s)
- Lorenzo Zaffiri
- 1 Department of Public Health and Infectious Disease, University of Rome "Sapienza," Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Abstract
PURPOSE OF REVIEW To discuss the recent major advances in the understanding of how host immune defenses contribute to HIV reservoir control. RECENT FINDINGS Immune control of HIV-1 reservoirs is a two-step process: viral replication activation from latent reservoirs followed by elimination of virus-expressing cells by the host. Environmental factors, such as pro-inflammatory type-I interferon, chemokines or cytokines, can facilitate HIV-1 replication, confer dormancy in CD4 cells or confer resistance to cytopathogenic effects of cytotoxic CD8 T cells. Therefore, they constitute a double-edged sword for immune control of HIV reservoirs. Concomitantly, adaptive immunity takes advantage of CD4 T-cell homeostatic mechanisms and can expose HIV-1 antigen-expressing cells to HIV-specific cytotoxic CD8 T cells, and limit virus spreading. These highly interconnected phenomena can lead to quasi-equilibrium between the HIV-1 reservoirs and host immune control that can serve as a model for the 'shock and kill' immune-based therapeutic strategies in play in the course of finding an HIV cure. SUMMARY Immune control of HIV reservoirs in CD4 T cells involves modulation of both HIV-1 latency and the continuous reseeding of the reservoir offering conceptual models that may advance HIV cure strategies.
Collapse
|
96
|
The role of T helper (TH)17 cells as a double-edged sword in the interplay of infection and autoimmunity with a focus on xenobiotic-induced immunomodulation. Clin Dev Immunol 2013; 2013:374769. [PMID: 24151516 PMCID: PMC3787652 DOI: 10.1155/2013/374769] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/04/2013] [Accepted: 06/22/2013] [Indexed: 12/18/2022]
Abstract
Extensive research in recent years suggests that exposure to xenobiotic stimuli plays a critical role in autoimmunity induction and severity and that the resulting response would be exacerbated in individuals with an infection-aroused immune system. In this context, heavy metals constitute a prominent category of xenobiotic substances, known to alter divergent immune cell responses in accidentally and occupationally exposed individuals, thereby increasing the susceptibility to autoimmunity and cancer, especially when accompanied by inflammation-triggered persistent sensitization. This perception is learned from experimental models of infection and epidemiologic studies and clearly underscores the interplay of exposure to such immunomodulatory elements with pre- or postexposure infectious events. Further, the TH17 cell subset, known to be associated with a growing list of autoimmune manifestations, may be the “superstar” at the interface of xenobiotic exposure and autoimmunity. In this review, the most recently established links to this nomination are short-listed to create a framework to better understand new insights into TH17's contributions to autoimmunity.
Collapse
|
97
|
Kim CJ, McKinnon LR, Kovacs C, Kandel G, Huibner S, Chege D, Shahabi K, Benko E, Loutfy M, Ostrowski M, Kaul R. Mucosal Th17 cell function is altered during HIV infection and is an independent predictor of systemic immune activation. THE JOURNAL OF IMMUNOLOGY 2013; 191:2164-73. [PMID: 23894197 DOI: 10.4049/jimmunol.1300829] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Mucosal Th17 cells maintain the gut epithelial barrier and prevent invasion by luminal bacteria through a delicate balance of immunosuppressive and proinflammatory functions. HIV infection is characterized by mucosal Th17 depletion, microbial translocation, and immune activation. Therefore, we assessed the function of blood and sigmoid Th17 cells during both early and chronic HIV infection, as well as the impact of short- and long-term antiretroviral therapy. Th17 cells were defined as IL-17a(+) CD4 T cells, and their functional capacity was assessed by the coproduction of the inflammatory cytokines IL-22, TNF-α, and IFN-γ, as well as the immunoregulatory cytokine IL-10. Gut Th17 cells had a much greater capacity to produce proinflammatory cytokines than did those from the blood, but this capacity was dramatically reduced from the earliest stages of HIV infection. Immunoregulatory skewing of mucosal Th17 cell function, characterized by an increased IL-10/TNF-α ratio, was uniquely seen during early HIV infection and was independently associated with reduced systemic immune activation. Antiretroviral therapy rapidly restored mucosal Th17 cell numbers; however, normalization of mucosal Th17 function, microbial translocation, and mucosal/systemic immune activation was much delayed. These findings emphasize that strategies to preserve or to more rapidly restore mucosal Th17 function may have important therapeutic benefit.
Collapse
Affiliation(s)
- Connie J Kim
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Brenchley JM. Mucosal immunity in human and simian immunodeficiency lentivirus infections. Mucosal Immunol 2013; 6:657-65. [PMID: 23549448 PMCID: PMC4154146 DOI: 10.1038/mi.2013.15] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overwhelming evidence indicates that distinct pathological phenomenon occurs within the gastrointestinal (GI) tract of progressively simian immunodeficiency virus (SIV)-infected Asian macaques and HIV-infected humans compared with other anatomical sites. Massive loss of GI tract lamina propria CD4 T cells, alteration in the profile of lymphocytic cytokine production, changes in the landscape of GI tract antigen-presenting cells, and variations to the structural barrier of the GI tract are hallmarks of progressive HIV/SIV infections. The pathology within the GI tract results in translocation of microbial products from the lumen of the intestine into peripheral circulation. These translocated microbial products directly stimulate the immune system and exacerbate immune activation and, thus, disease progression. Initiation of combination antiretroviral therapy (cART) does not restore completely the immunological abnormalities within the GI tract. This incomplete restoration within the GI tract may contribute to the increased mortality observed within HIV-infected individuals treated for decades with cART. Novel therapeutic interventions aimed at enhancing GI tract anatomy and physiology may improve the prognosis of HIV-infected individuals.
Collapse
Affiliation(s)
- JM Brenchley
- Program in Tissue Immunity and Repair and Lab of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
99
|
Chevalier MF, Petitjean G, Dunyach-Rémy C, Didier C, Girard PM, Manea ME, Campa P, Meyer L, Rouzioux C, Lavigne JP, Barré-Sinoussi F, Scott-Algara D, Weiss L. The Th17/Treg ratio, IL-1RA and sCD14 levels in primary HIV infection predict the T-cell activation set point in the absence of systemic microbial translocation. PLoS Pathog 2013; 9:e1003453. [PMID: 23818854 PMCID: PMC3688532 DOI: 10.1371/journal.ppat.1003453] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/08/2013] [Indexed: 12/31/2022] Open
Abstract
Impairment of the intestinal barrier and subsequent microbial translocation (MT) may be involved in chronic immune activation, which plays a central role in HIV pathogenesis. Th17 cells are critical to prevent MT. The aim of the study was to investigate, in patients with primary HIV infection (PHI), the early relationship between the Th17/Treg ratio, monocyte activation and MT and their impact on the T-cell activation set point, which is known to predict disease progression. 27 patients with early PHI were included in a prospective longitudinal study and followed-up for 6 months. At baseline, the Th17/Treg ratio strongly negatively correlated with the proportion of activated CD8 T cells expressing CD38/HLA-DR or Ki-67. Also, the Th17/Treg ratio was negatively related to viral load and plasma levels of sCD14 and IL-1RA, two markers of monocyte activation. In untreated patients, the Th17/Treg ratio at baseline negatively correlated with CD8 T-cell activation at month 6 defining the T-cell activation set point (% HLA-DR(+)CD38(+) and %Ki-67(+)). Soluble CD14 and IL-1RA plasma levels also predicted the T-cell activation set point. Levels of I-FABP, a marker of mucosal damages, were similar to healthy controls at baseline but increased at month 6. No decrease in anti-endotoxin core antibody (EndoCAb) and no peptidoglycan were detected during PHI. In addition, 16S rDNA was only detected at low levels in 2 out 27 patients at baseline and in one additional patient at M6. Altogether, data support the hypothesis that T-cell and monocyte activation in PHI are not primarily driven by systemic MT but rather by viral replication. Moreover, the "innate immune set point" defined by the early levels of sCD14 and IL-1RA might be powerful early surrogate markers for disease progression and should be considered for use in clinical practice.
Collapse
Affiliation(s)
- Mathieu F. Chevalier
- Institut Pasteur, Régulation des infections rétrovirales, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Gaël Petitjean
- Institut Pasteur, Régulation des infections rétrovirales, Paris, France
| | - Catherine Dunyach-Rémy
- INSERM U1047, Université Montpellier 1, UFR de Médecine, Nîmes, France
- Laboratoire de Bactériologie, CHU Carémeau, Nîmes, France
| | - Céline Didier
- Institut Pasteur, Régulation des infections rétrovirales, Paris, France
| | | | | | | | - Laurence Meyer
- INSERM U 1018, AP-HP, Université Paris Sud, Paris, France
| | - Christine Rouzioux
- AP-HP, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Philippe Lavigne
- INSERM U1047, Université Montpellier 1, UFR de Médecine, Nîmes, France
- Laboratoire de Bactériologie, CHU Carémeau, Nîmes, France
| | | | | | - Laurence Weiss
- Institut Pasteur, Régulation des infections rétrovirales, Paris, France
- AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
100
|
Abstract
In pathogenic simian immunodeficiency virus (SIV) and human immunodeficiency virus (HIV) infections, the translocation of microbial products from the gastrointestinal (GI) tract to portal and systemic circulation has been proposed as a major driver of the chronic immune activation that is associated with disease progression. Consistently, microbial translocation is not present in nonpathogenic SIV infections of natural host species. In vivo studies demonstrated that HIV/SIV-associated microbial translocation results from a series of immunopathological events occurring at the GI mucosa: (i) early and severe mucosal CD4(+) depletion, (ii) mucosal immune hyperactivation/persistent inflammation; (iii) damage to the integrity of the intestinal epithelium with enterocyte apoptosis and tight junction disruption; and (iv) subverted the gut microbiome, with a predominance of opportunistic bacteria. Direct in situ evidence of microbial translocation has been provided for SIV-infected rhesus macaques showing translocated microbial products in the intestinal lamina propria and distant sites. While the mechanisms by which microbial translocation causes immune activation remain controversial, a key pathogenic event appears to be innate immunity activation via Toll-like receptors and other pathogen recognition receptors. Accumulating clinical observations suggest that microbial translocation might affect HIV disease progression, response to therapy, and non-AIDS comorbidities. Given its detrimental effect on overall immunity, several interventions to prevent/block microbial translocation are currently under investigation as novel therapeutic agents for HIV/AIDS.
Collapse
|