1
|
Vellas C, Nayrac M, Collercandy N, Requena M, Jeanne N, Latour J, Dimeglio C, Cazabat M, Barange K, Alric L, Carrere N, Martin-Blondel G, Izopet J, Delobel P. Intact proviruses are enriched in the colon and associated with PD-1 +TIGIT - mucosal CD4 + T cells of people with HIV-1 on antiretroviral therapy. EBioMedicine 2024; 100:104954. [PMID: 38160480 PMCID: PMC10792747 DOI: 10.1016/j.ebiom.2023.104954] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND The persistence of intact replication-competent HIV-1 proviruses is responsible for the virological rebound off treatment. The gut could be a major reservoir of HIV-1 due to the high number of infected target cells. METHODS We collected blood samples and intestinal biopsies (duodenum, ileum, colon) from 42 people with HIV-1 receiving effective antiretroviral therapy. We used the Intact Proviral DNA Assay to estimate the frequency of intact HIV-1 proviruses in the blood and in the intestinal mucosa of these individuals. We analyzed the genetic complexity of the HIV-1 reservoir by performing single-molecule next-generation sequencing of HIV-1 env DNA. The activation/exhaustion profile of mucosal T lymphocytes was assessed by flow cytometry. FINDINGS Intact proviruses are particularly enriched in the colon. Residual HIV-1 transcription in the gut is associated with persistent mucosal and systemic immune activation. The HIV-1 intestinal reservoir appears to be shaped by the proliferation of provirus-hosting cells. The genetic complexity of the viral reservoir in the colon is positively associated with TIGIT expression but negatively with PD-1, and inversely related to its intact content. The size of the intact reservoir in the colon is associated with PD-1+TIGIT- mucosal CD4+ T cells, particularly in CD27+ memory cells, whose proliferation and survival could contribute to the enrichment of the viral reservoir by intact proviruses. INTERPRETATION Enrichment in intact proviruses makes the gut a key compartment for HIV-1 persistence on antiretroviral therapy. FUNDING This project was supported by grants from the ANRS-MIE (ANRS EP61 GALT), Sidaction, and the Institut Universitaire de France.
Collapse
Affiliation(s)
- Camille Vellas
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France
| | - Manon Nayrac
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France
| | - Nived Collercandy
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France; CHU de Toulouse, Service des Maladies Infectieuses et Tropicales, Toulouse F-31300, France
| | - Mary Requena
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France; CHU de Toulouse, Laboratoire de Virologie, Toulouse F-31300, France
| | - Nicolas Jeanne
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France; CHU de Toulouse, Laboratoire de Virologie, Toulouse F-31300, France
| | - Justine Latour
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France; CHU de Toulouse, Laboratoire de Virologie, Toulouse F-31300, France
| | - Chloé Dimeglio
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France
| | - Michelle Cazabat
- CHU de Toulouse, Laboratoire de Virologie, Toulouse F-31300, France
| | - Karl Barange
- CHU de Toulouse, Service d'Hépato-Gastro-Entérologie, Toulouse F-31400, France
| | - Laurent Alric
- Université Toulouse III Paul Sabatier, Toulouse F-31400, France; CHU de Toulouse, Service de Médecine Interne et Immunologie clinique, Toulouse F-31400, France
| | - Nicolas Carrere
- Université Toulouse III Paul Sabatier, Toulouse F-31400, France; CHU de Toulouse, Service de Chirurgie Générale et Digestive, Toulouse F-31400, France
| | - Guillaume Martin-Blondel
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France; CHU de Toulouse, Service des Maladies Infectieuses et Tropicales, Toulouse F-31300, France; Université Toulouse III Paul Sabatier, Toulouse F-31400, France
| | - Jacques Izopet
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France; CHU de Toulouse, Laboratoire de Virologie, Toulouse F-31300, France; Université Toulouse III Paul Sabatier, Toulouse F-31400, France
| | - Pierre Delobel
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse F-31300, France; CHU de Toulouse, Service des Maladies Infectieuses et Tropicales, Toulouse F-31300, France; Université Toulouse III Paul Sabatier, Toulouse F-31400, France.
| |
Collapse
|
2
|
Guo YT, Guo XY, Fan LN, Wang ZR, Qu MM, Zhang C, Fan X, Song JW, Yang BP, Zhang JY, Xu R, Jiao YM, Ma P, Chen YK, Wang FS. The Imbalance Between Intestinal Th17 and Treg Cells Is Associated with an Incomplete Immune Reconstitution During Long-Term Antiretroviral Therapy in Patients with HIV. Viral Immunol 2023; 36:331-342. [PMID: 37184871 DOI: 10.1089/vim.2023.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Studies assessing the gut mucosal immune balance in HIV-infected patients using intestinal samples are scarce. In this study, we used intestinal mucosal specimens from the ileocecal region of seven immunological nonresponders (INRs), nine immunological responders (IRs), and six HIV-negative controls. We investigated T helper 17 (Th17) and T regulatory (Treg) cell counts and their ratio, zonula occludens-1 (ZO-1), intestinal fatty acid-binding protein (I-FABP), tumor necrosis factor-α, CD4+ T cell counts, HIV DNA, and cell-associated HIV RNA. The results showed that INRs had lower Th17 and higher Treg cell counts than IR, resulting in a significant difference in the Th17/Treg ratio between IRs and INRs. In addition, INRs had lower ZO-1 and higher I-FABP levels than IRs. The Th17/Treg ratio was positively associated with ZO-1 and negatively associated with I-FABP levels. There was a positive correlation between Th17/Treg ratio and CD4+ T cell counts and a negative correlation between the Th17/Treg ratio and HIV DNA in the intestine. Our study suggests that the imbalance of Th17/Treg in the intestine is a characteristic of incomplete immune reconstitution to antiretroviral therapy and is associated with intestinal damage.
Collapse
Affiliation(s)
- Yun-Tian Guo
- Department of Internal Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xiao-Yan Guo
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Li-Na Fan
- Department of Infectious Diseases, Tianjin Second People's Hospital, Tianjin, China
| | - Ze-Rui Wang
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Meng-Meng Qu
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xing Fan
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Wen Song
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Bao-Peng Yang
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ji-Yuan Zhang
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ruonan Xu
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yan-Mei Jiao
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ping Ma
- Department of Infectious Diseases, Tianjin Second People's Hospital, Tianjin, China
| | - Yao-Kai Chen
- Department of Infectious Disease, Chongqing Public Health Medical Center, Chongqing, China
| | - Fu-Sheng Wang
- Department of Internal Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Senior Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| |
Collapse
|
3
|
Petkov S, Chiodi F. Impaired CD4+ T cell differentiation in HIV-1 infected patients receiving early anti-retroviral therapy. Genomics 2022; 114:110367. [PMID: 35429609 DOI: 10.1016/j.ygeno.2022.110367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/01/2022] [Accepted: 04/09/2022] [Indexed: 01/14/2023]
Abstract
Differentiation of CD4+ T naïve (TN) into central memory (TCM) cells involves extensive molecular processes. We compared the transcriptomes of CD4+ TN and TCM cells from HIV-1 infected patients receiving early anti-retroviral therapy (ART; EA; n = 13) and controls (n = 15). Comparison of protein coding genes between TCM and TN revealed 533 and 82 differentially expressed genes (DEGs) in controls and EA, respectively. A high degree of transcriptional complexity was detected during transition of CD4+ TN to TCM cells in controls involving 70 TFs, 20 master regulators of T cell differentiation (TBX21, GATA3, RARA, FOXP3, RORC); in EA only 7 TFs were modulated with expression of several master regulators remaining unchanged during differentiation. Analysis of interactions between modulated TFs and target genes revealed important regulatory interactions missing in EA group. We conclude that T cell differentiation in EA patients is impaired due to reduced modulation of genes involved in transition from CD4+ TN to TCM cells.
Collapse
Affiliation(s)
- Stefan Petkov
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Solna, Sweden
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
4
|
Impact of Early ARV Initiation on Relative Proportions of Effector and Regulatory CD8 T Cell in Mesenteric Lymph Nodes and Peripheral Blood During Acute SIV Infection of Rhesus Macaques. J Virol 2022; 96:e0025522. [PMID: 35311550 PMCID: PMC9006892 DOI: 10.1128/jvi.00255-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
CD8 T cells are key players in the clearance of human immunodeficiency virus (HIV)-infected cells, such that CD8 T-cell dysfunction contributes to viral persistence despite antiretroviral (ARV) therapy. Mesenteric lymph nodes (MLNs) are major sites of gut mucosal immunity. While different CD8 T cell subsets such as CD8 alpha-alpha (CD8αα), CD8 alpha-beta (CD8αβ), CD8 regulatory T cells (Treg), and mucosa-associated invariant T cells (MAIT) are present in the gut and exhibit distinct functions, their dynamics remain poorly understood due to the lack of accessibility to these tissues in humans. We thus assessed CD8 T cells in MLNs versus peripheral blood in simian immunodeficiency virus (SIV)-infected rhesus macaques (RMs) following early ARV therapy initiation. SIV infection was associated with an increase over time of both CD8αβ and CD8αα T cells in the blood and MLNs, whereas early ARV initiation significantly decreased the frequencies of CD8αα but not CD8αβ T cells in MLNs. A significant decrease in the expression of chemokine receptors CCR6 and CXCR3 by CD8 T cells, which are essential for T-cell trafficking to the inflammatory sites, was observed in chronically SIV-infected RMs. Surprisingly, while MAIT cells are increased in ARV-treated RMs, their frequencies in MLN are extremely low and were not impacted by ARV. The acute infection resulted in an early CD39+FoxP3+ CD8 Tregs increase in both compartments, which was normalized after early ARV. Frequencies of CD8 Treg cells were positively correlated with frequencies of CD4 Tregs and accordingly negatively correlated with the Th17/Treg ratio in the blood but not in MLNs. Overall, our results underscore the difference in CD8 T-cell subset dynamics in the blood and MLNs. IMPORTANCE Changes in CD8 T-cell subsets during acute SIV/HIV infections and following early ARV initiation in gut lymphoid tissues are poorly understood. Using an acute SIV infection model in rhesus macaques, we assessed the impact of early ARV, initiated 4 days postinfection, on relative proportions of CD8 T-cell subsets in MLNs compared to blood. We found that acute SIV infection and early ARV initiation differentially affect the distribution of effector CD8 T cells, CD8 MAIT cells, and CD8 Tregs in MLNs compared to blood. Overall, early ARV initiation maintains the frequency of effector CD8 T cells while reducing immunosuppressive CD39+ CD8 Tregs. Our study provides deeper insight into the dynamics of the CD8 T-cell compartment in gut mucosal immune surveillance during acute SIV infection and following early ARV initiation.
Collapse
|
5
|
Zhang F, Sun L, Lafferty MK, Margolick JB, Garzino-Demo A. Decreased MIP-3α Production from Antigen-Activated PBMCs in Symptomatic HIV-Infected Subjects. Pathogens 2021; 11:pathogens11010007. [PMID: 35055955 PMCID: PMC8778881 DOI: 10.3390/pathogens11010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 11/30/2022] Open
Abstract
CD4+ CCR6+ T cells are highly susceptible to HIV infection, and a high cytokine producing CCR6+ T cell subset is selectively lost during HIV infection. The CCR6 chemokine MIP-3α (CCL20) is produced at sites of infection in SIV animal models. Recently, we have shown that MIP-3α inhibits HIV replication. This inhibition of HIV infection is mediated by CCR6 signaling and eventuates in increased APOBEC3G expression. Since there are few existing reports on the role of MIP-3α in health or disease, we studied its production by PBMCs from HIV-seronegative and HIV+ subjects. We evaluated the ability of PBMCs to produce MIP-3α in response to antigen stimulation using cells obtained from two groups: one composed of HIV-seronegative subjects (n = 16) and the other composed of HIV+ subjects (n = 58), some asymptomatic and some with clinically defined AIDS. Antigens included fragment C of the tetanus toxin, Candida albicans, whole-inactivated HIV, and HIV p24. MIP-3α was detected by ELISA in tissue culture supernatants of antigen-stimulated PBMCs. MIP-3α production by antigen-stimulated PBMCs was readily measured for HIV-negative subjects and for HIV-seropositive asymptomatic subjects, but not for patients with AIDS. These results suggest that subversion of the MIP-3α-CCR6 axis by HIV during the course of infection contributes to the loss of immune function that eventually leads to AIDS.
Collapse
Affiliation(s)
- Fuchun Zhang
- Laboratory of Virus-Host Interactions, Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Department of Microbiology and Immunology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA; (F.Z.); (L.S.); (M.K.L.)
- Department of Infectious Diseases, Guangzhou No. 8 People’s Hospital, Guangzhou Medical College, Guangzhou 510060, China
| | - Lingling Sun
- Laboratory of Virus-Host Interactions, Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Department of Microbiology and Immunology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA; (F.Z.); (L.S.); (M.K.L.)
| | - Mark K. Lafferty
- Laboratory of Virus-Host Interactions, Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Department of Microbiology and Immunology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA; (F.Z.); (L.S.); (M.K.L.)
| | - Joseph B. Margolick
- Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA;
| | - Alfredo Garzino-Demo
- Laboratory of Virus-Host Interactions, Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Department of Microbiology and Immunology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA; (F.Z.); (L.S.); (M.K.L.)
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy
- Correspondence: or
| |
Collapse
|
6
|
Wiche Salinas TR, Gosselin A, Raymond Marchand L, Moreira Gabriel E, Tastet O, Goulet JP, Zhang Y, Vlad D, Touil H, Routy JP, Bego MG, El-Far M, Chomont N, Landay AL, Cohen ÉA, Tremblay C, Ancuta P. IL-17A reprograms intestinal epithelial cells to facilitate HIV-1 replication and outgrowth in CD4+ T cells. iScience 2021; 24:103225. [PMID: 34712922 PMCID: PMC8531570 DOI: 10.1016/j.isci.2021.103225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 08/09/2021] [Accepted: 10/01/2021] [Indexed: 12/25/2022] Open
Abstract
The crosstalk between intestinal epithelial cells (IECs) and Th17-polarized CD4+ T cells is critical for mucosal homeostasis, with HIV-1 causing significant alterations in people living with HIV (PLWH) despite antiretroviral therapy (ART). In a model of IEC and T cell co-cultures, we investigated the effects of IL-17A, the Th17 hallmark cytokine, on IEC ability to promote de novo HIV infection and viral reservoir reactivation. Our results demonstrate that IL-17A acts in synergy with TNF to boost IEC production of CCL20, a Th17-attractant chemokine, and promote HIV trans-infection of CD4+ T cells and viral outgrowth from reservoir cells of ART-treated PLWH. Importantly, the Illumina RNA-sequencing revealed an IL-17A-mediated pro-inflammatory and pro-viral molecular signature, including a decreased expression of type I interferon (IFN-I)-induced HIV restriction factors. These findings point to the deleterious features of IL-17A and raise awareness for caution when designing therapies aimed at restoring the paucity of mucosal Th17 cells in ART-treated PLWH. IL-17A acts in synergy with TNF to enhance CCL20 production in IEC exposed to HIV IL-17A/TNF-activated IEC efficiently promote HIV trans-infection of CD4+ T cells IL-17A reprograms IEC to boost HIV outgrowth from CD4+ T cells of ART-treated PLWH IL-17A decreases the expression of IFN-I-induced HIV restriction factors in IEC
Collapse
Affiliation(s)
- Tomas Raul Wiche Salinas
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Annie Gosselin
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | | | - Etiene Moreira Gabriel
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Olivier Tastet
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | | | - Yuwei Zhang
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | - Dragos Vlad
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | - Hanane Touil
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Division of Hematology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Mariana G. Bego
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
| | - Mohamed El-Far
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | - Nicolas Chomont
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Éric A. Cohen
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
| | - Cécile Tremblay
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Petronela Ancuta
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- Corresponding author
| |
Collapse
|
7
|
Yero A, Shi T, Farnos O, Routy JP, Tremblay C, Durand M, Tsoukas C, Costiniuk CT, Jenabian MA. Dynamics and epigenetic signature of regulatory T-cells following antiretroviral therapy initiation in acute HIV infection. EBioMedicine 2021; 71:103570. [PMID: 34500304 PMCID: PMC8429924 DOI: 10.1016/j.ebiom.2021.103570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND HIV infection promotes the expansion of immunosuppressive regulatory T-cells (Tregs), contributing to immune dysfunction, tissue fibrosis and disease progression. Early antiretroviral treatment (ART) upon HIV infection improves CD4 count and decreases immune activation. However, Treg dynamics and their epigenetic regulation following early ART initiation remain understudied. METHODS Treg subsets were characterized by flow cytometry in 103 individuals, including untreated HIV-infected participants in acute and chronic phases, ART-treated in early infection, elite controllers (ECs), immunological controllers (ICs), and HIV-uninfected controls. The methylation status of six regulatory regions of the foxp3 gene was assessed using MiSeq technology. FINDINGS Total Treg frequency increased overtime during HIV infection, which was normalized in early ART recipients. Tregs in untreated individuals expressed higher levels of activation and immunosuppressive markers (CD39, and LAP(TGF-β1)), which remained unchanged following early ART. Expression of gut migration markers (CCR9, Integrin-β7) by Tregs was elevated during untreated HIV infection, while they declined with the duration of ART but not upon early ART initiation. Notably, gut-homing Tregs expressing LAP(TGF-β1) and CD39 remained higher despite early treatment. Additionally, the increase in LAP(TGF-β1)+ Tregs overtime were consistent with higher demethylation of conserved non-coding sequence (CNS)-1 in the foxp3 gene. Remarkably, LAP(TGF-β1)-expressing Tregs in ECs were significantly higher than in uninfected subjects, while the markers of Treg activation and gut migration were not different. INTERPRETATION Early ART initiation was unable to control the levels of immunosuppressive Treg subsets and their gut migration potential, which could ultimately contribute to gut tissue fibrosis and HIV disease progression. FUNDING This study was funded by the Canadian Institutes of Health Research (CIHR, grant MOP 142294) and in part by the AIDS and Infectious Diseases Network of the Réseau SIDA et maladies infectieuses du Fonds de recherche du Québec-Santé (FRQ-S).
Collapse
Affiliation(s)
- Alexis Yero
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - Tao Shi
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - Omar Farnos
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - Jean-Pierre Routy
- Research Institute of McGill University Health Centre, Montreal, QC, Canada; Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, Glen Site, McGill University Health Centre, Montreal, QC, Canada
| | - Cécile Tremblay
- CHUM Research Centre, Montreal, QC, Canada; Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | | | - Christos Tsoukas
- Research Institute of McGill University Health Centre, Montreal, QC, Canada; Division of Clinical Immunology and Allergy, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Cecilia T Costiniuk
- Research Institute of McGill University Health Centre, Montreal, QC, Canada; Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada; Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada; Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
8
|
Hou X, Zhu F, Ni Y, Chen T, Du J, Liu X, Han Y, Liu Y, Du W, Li Y, Wang X, Li D, Liang R, Li B, Shi G. USP4 is pathogenic in allergic airway inflammation by inhibiting regulatory T cell response. Life Sci 2021; 281:119720. [PMID: 34144056 DOI: 10.1016/j.lfs.2021.119720] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/05/2021] [Accepted: 06/05/2021] [Indexed: 11/17/2022]
Abstract
AIMS Asthma is characterized by chronic inflammation and airway hyperresponsiveness (AHR). It is controllable, but not curable. Ubiquitin-specific peptidase 4 (USP4) has been verified as a regulator of regulatory T (Treg) cells and Th17 cells in vitro. In this study, we aim to investigate whether USP4 could serve as a therapeutic target for asthma. MAIN METHODS Age-matched USP4 wild-type and knockout mice received an intraperitoneal injection of 100 μg ovalbumin (OVA) mixed in 2 mg aluminum hydroxide in 1 × PBS on days 0, 7 and 14. On days 21 to 27, the mice were challenged with aerosolized 1% OVA in 1 × PBS for 30 min. Tissue histology, ELISA and flow cytometry were applied 24 h after the last OVA challenge. KEY FINDINGS USP4 deficiency protected mice from OVA-induced AHR and decreased the production of several inflammatory cytokines in T cells in vivo. Compared to the lung cells isolated from WT mice, Usp4-/- lung cells decreased secretion of IL-4, IL-13 and IL-17A upon stimulation in vitro. Meanwhile, the percentage of CD4+Foxp3+ Treg cells was elevated, with more CCR6+Foxp3+ Treg cells accumulating in the lungs of OVA-challenged USP4 deficient mice than in their wild-type counterparts. Treatment with the USP4 inhibitor, Vialinin A, reduced inflammatory cell infiltration in the lungs of OVA-challenged mice in vivo. SIGNIFICANCE We found USP4 deficiency contributes to attenuated airway inflammation and AHR in allergen-induced murine asthma, and Vialinin A treatment alleviates asthma pathogenesis and may serve as a promising therapeutic target for asthma.
Collapse
Affiliation(s)
- Xiaoxia Hou
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangming Zhu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Institute Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai, China
| | - Yingmeng Ni
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiantian Chen
- Department of Pulmonary and Critical Care Medicine, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Du
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinnan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yichao Han
- Department of Thoracic Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yahui Liu
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Du
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxia Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Liang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Guochao Shi
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Ogongo P, Tezera LB, Ardain A, Nhamoyebonde S, Ramsuran D, Singh A, Ng’oepe A, Karim F, Naidoo T, Khan K, Dullabh KJ, Fehlings M, Lee BH, Nardin A, Lindestam Arlehamn CS, Sette A, Behar SM, Steyn AJ, Madansein R, Kløverpris HN, Elkington PT, Leslie A. Tissue-resident-like CD4+ T cells secreting IL-17 control Mycobacterium tuberculosis in the human lung. J Clin Invest 2021; 131:142014. [PMID: 33848273 PMCID: PMC8121523 DOI: 10.1172/jci142014] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
T cell immunity is essential for the control of tuberculosis (TB), an important disease of the lung, and is generally studied in humans using peripheral blood cells. Mounting evidence, however, indicates that tissue-resident memory T cells (Trms) are superior at controlling many pathogens, including Mycobacterium tuberculosis (M. tuberculosis), and can be quite different from those in circulation. Using freshly resected lung tissue, from individuals with active or previous TB, we identified distinct CD4+ and CD8+ Trm-like clusters within TB-diseased lung tissue that were functional and enriched for IL-17-producing cells. M. tuberculosis-specific CD4+ T cells producing TNF-α, IL-2, and IL-17 were highly expanded in the lung compared with matched blood samples, in which IL-17+ cells were largely absent. Strikingly, the frequency of M. tuberculosis-specific lung T cells making IL-17, but not other cytokines, inversely correlated with the plasma IL-1β levels, suggesting a potential link with disease severity. Using a human granuloma model, we showed the addition of either exogenous IL-17 or IL-2 enhanced immune control of M. tuberculosis and was associated with increased NO production. Taken together, these data support an important role for M. tuberculosis-specific Trm-like, IL-17-producing cells in the immune control of M. tuberculosis in the human lung.
Collapse
Affiliation(s)
- Paul Ogongo
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Institute of Primate Research, National Museums of Kenya, Nairobi, Kenya
| | - Liku B. Tezera
- National Institute for Health Research Southampton Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, and
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Amanda Ardain
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Shepherd Nhamoyebonde
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | - Alveera Singh
- Africa Health Research Institute, Durban, South Africa
| | | | - Farina Karim
- Africa Health Research Institute, Durban, South Africa
| | - Taryn Naidoo
- Africa Health Research Institute, Durban, South Africa
| | - Khadija Khan
- Africa Health Research Institute, Durban, South Africa
| | - Kaylesh J. Dullabh
- Department of Cardiothoracic Surgery, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | | | | | | | - Alessandro Sette
- La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Adrie J.C. Steyn
- Africa Health Research Institute, Durban, South Africa
- Department of Microbiology and
- Center for AIDS Research and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rajhmun Madansein
- Department of Cardiothoracic Surgery, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Henrik N. Kløverpris
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Paul T. Elkington
- National Institute for Health Research Southampton Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, and
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
10
|
Blondin-Ladrie L, Aranguren M, Doyon-Laliberté K, Poudrier J, Roger M. The Importance of Regulation in Natural Immunity to HIV. Vaccines (Basel) 2021; 9:vaccines9030271. [PMID: 33803543 PMCID: PMC8003059 DOI: 10.3390/vaccines9030271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Worldwide, most Human Immunodeficiency Virus (HIV) infections are acquired through heterosexual intercourse, and in sub-Saharan Africa, 59% of new HIV infections affect women. Vaccines and microbicides hold promise for preventing the acquisition of HIV. To this end, the study of HIV highly exposed seronegative (HESN) female commercial sex workers (CSWs), who constitute a model of natural immunity to HIV, provides an exceptional opportunity to determine important clues for the development of preventive strategies. Studies using both female genital tract (FGT) and peripheral blood samples of HESN CSWs, have allowed identifying distinct features, notably low-inflammatory patterns associated with resistance to infection. How this seemingly regulated response is achieved at the initial site of HIV infection remains unknown. One hypothesis is that populations presenting regulatory profiles contribute to the orchestration of potent anti-viral and low-inflammatory responses at the initial site of HIV transmission. Here, we view to update our knowledge regarding this issue.
Collapse
Affiliation(s)
- Laurence Blondin-Ladrie
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Matheus Aranguren
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Kim Doyon-Laliberté
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Johanne Poudrier
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
- Correspondence: (J.P.); (M.R.)
| | - Michel Roger
- Axe Immunopathologie, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X0A9, Canada; (L.B.-L.); (M.A.); (K.D.-L.)
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3C3J7, Canada
- Institut National de Santé Publique du Québec, Montréal, QC H2P1E2, Canada
- Correspondence: (J.P.); (M.R.)
| |
Collapse
|
11
|
O’Neil TR, Hu K, Truong NR, Arshad S, Shacklett BL, Cunningham AL, Nasr N. The Role of Tissue Resident Memory CD4 T Cells in Herpes Simplex Viral and HIV Infection. Viruses 2021; 13:359. [PMID: 33668777 PMCID: PMC7996247 DOI: 10.3390/v13030359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Tissue-resident memory T cells (TRM) were first described in 2009. While initially the major focus was on CD8+ TRM, there has recently been increased interest in defining the phenotype and the role of CD4+ TRM in diseases. Circulating CD4+ T cells seed CD4+ TRM, but there also appears to be an equilibrium between CD4+ TRM and blood CD4+ T cells. CD4+ TRM are more mobile than CD8+ TRM, usually localized deeper within the dermis/lamina propria and yet may exhibit synergy with CD8+ TRM in disease control. This has been demonstrated in herpes simplex infections in mice. In human recurrent herpes infections, both CD4+ and CD8+ TRM persisting between lesions may control asymptomatic shedding through interferon-gamma secretion, although this has been more clearly shown for CD8+ T cells. The exact role of the CD4+/CD8+ TRM axis in the trigeminal ganglia and/or cornea in controlling recurrent herpetic keratitis is unknown. In HIV, CD4+ TRM have now been shown to be a major target for productive and latent infection in the cervix. In HSV and HIV co-infections, CD4+ TRM persisting in the dermis support HIV replication. Further understanding of the role of CD4+ TRM and their induction by vaccines may help control sexual transmission by both viruses.
Collapse
Affiliation(s)
- Thomas R. O’Neil
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Kevin Hu
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Naomi R. Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Sana Arshad
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616, USA;
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia
| |
Collapse
|
12
|
Th22 cells are efficiently recruited in the gut by CCL28 as an alternative to CCL20 but do not compensate for the loss of Th17 cells in treated HIV-1-infected individuals. Mucosal Immunol 2021; 14:219-228. [PMID: 32346082 DOI: 10.1038/s41385-020-0286-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 02/19/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
Gut CD4+ T cells are incompletely restored in most HIV-1-infected individuals on antiretroviral therapy, notably Th17 cells, a key subset in mucosal homeostasis. By contrast, gut Th22 cells are usually restored at normal frequencies. Th22 cells display a CCR6+CCR10+ phenotype and could thus respond to CCL20- and CCL28-mediated chemotaxis, while Th17 cells, which express CCR6 but not CCR10, depend on CCL20. Herein, we found that CCL28 is normally expressed by duodenal enterocytes of treated HIV-1-infected individuals, while CCL20 expression is blunted. Ex vivo, we showed that Th22 cells contribute to the reduction of CCL20 production by enterocytes through an IL-22- and IL-18-dependent mechanism. Th22 cells preferentially migrate via CCL20- rather than CCL28-mediated chemotaxis when both chemokines are available in the microenvironment. However, when the CCL20/CCL28 ratio drops, as in treated HIV-1-infected individuals, Th22 cells can migrate via the CCR10-CCL28 axis, as an alternative to CCR6-CCL20. This could explain the better reconstitution of gut Th22 compared with Th17 cells on antiretroviral therapy. Lastly, we assessed the relationships between the frequencies of gut Th17 and Th22 cells and inflammatory markers related to microbial translocation, and showed that Th22 cells do not compensate for the loss of Th17 cells in treated HIV-1-infected individuals.
Collapse
|
13
|
Anderson JL, Khoury G, Fromentin R, Solomon A, Chomont N, Sinclair E, Milush JM, Hartogensis W, Bacchetti P, Roche M, Tumpach C, Gartner M, Pitman MC, Epling CL, Hoh R, Hecht FM, Somsouk M, Cameron PU, Deeks SG, Lewin SR. Human Immunodeficiency Virus (HIV)-Infected CCR6+ Rectal CD4+ T Cells and HIV Persistence On Antiretroviral Therapy. J Infect Dis 2020; 221:744-755. [PMID: 31796951 PMCID: PMC7026892 DOI: 10.1093/infdis/jiz509] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023] Open
Abstract
Background Identifying where human immunodeficiency virus (HIV) persists in people living with HIV and receiving antiretroviral therapy is critical to develop cure strategies. We assessed the relationship of HIV persistence to expression of chemokine receptors and their chemokines in blood (n = 48) and in rectal (n = 20) and lymph node (LN; n = 8) tissue collected from people living with HIV who were receiving suppressive antiretroviral therapy. Methods Cell-associated integrated HIV DNA, unspliced HIV RNA, and chemokine messenger RNA were quantified by quantitative polymerase chain reaction. Chemokine receptor expression on CD4+ T cells was determined using flow cytometry. Results Integrated HIV DNA levels in CD4+ T cells, CCR6+CXCR3+ memory CD4+ T-cell frequency, and CCL20 expression (ligand for CCR6) were highest in rectal tissue, where HIV-infected CCR6+ T cells accounted for nearly all infected cells (median, 89.7%). Conversely in LN tissue, CCR6+ T cells were infrequent, and there was a statistically significant association of cell-associated HIV DNA and RNA with CCL19, CCL21, and CXCL13 chemokines. Conclusions HIV-infected CCR6+ CD4+ T cells accounted for the majority of infected cells in rectal tissue. The different relationships between HIV persistence and T-cell subsets and chemokines in rectal and LN tissue suggest that different tissue-specific strategies may be required to eliminate HIV persistence and that assessment of biomarkers for HIV persistence may not be generalizable between blood and other tissues.
Collapse
Affiliation(s)
- Jenny L Anderson
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Gabriela Khoury
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Rémi Fromentin
- Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Ajantha Solomon
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Nicolas Chomont
- Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Elizabeth Sinclair
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey M Milush
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Wendy Hartogensis
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Michael Roche
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Carolin Tumpach
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Matthew Gartner
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Matthew C Pitman
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Christine Lorrie Epling
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Rebecca Hoh
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Frederick M Hecht
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Ma Somsouk
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Paul U Cameron
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Steven G Deeks
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Marion O, Lhomme S, Nayrac M, Dubois M, Pucelle M, Requena M, Migueres M, Abravanel F, Peron JM, Carrere N, Suc B, Delobel P, Kamar N, Izopet J. Hepatitis E virus replication in human intestinal cells. Gut 2020; 69:901-910. [PMID: 31727684 DOI: 10.1136/gutjnl-2019-319004] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Hepatitis E virus (HEV), one of the most common agent of acute hepatitis worldwide, is mainly transmitted enterically, via contaminated water for HEV genotypes 1 (HEV1) and HEV2, or by eating raw or undercooked infected meat for HEV genotype 3 (HEV3) and HEV4. However, little is known about how the ingested HEV reaches the liver or its ability to replicate in intestinal cells. DESIGN We developed human primary cultures of small intestine epithelial cells and intestinal explants obtained from small bowel resections. The epithelial cells were also polarised on transwells. Cells were infected with Kernow-p6 strain or clinically derived virions. RESULTS Primary intestinal cells supported the growth of Kernow-p6 strain and HEV1 and HEV3 clinically derived virions. Polarised enterocytes infected with HEV1 and HEV3 strains released HEV particles vectorially: mostly into the apical compartment with a little basally. Iodixanol density gradient centrifugation of enterocyte-derived HEV virions gave bands at a density of 1.06-1.08 g/cm3, corresponding to that of quasi-enveloped HEV particles. Ribavirin therapy inhibited HEV excretion from the basal surface but not from the apical side of infected human enterocytes. HEV virions also infected intestinal tissue explants. Lastly, HEV RNA and antigen were detected in the intestinal crypts of a chronically infected patient. CONCLUSION HEV can replicate in intestinal cells and reaches the liver as quasi-enveloped virions.
Collapse
Affiliation(s)
- Olivier Marion
- Department of Nephrology and Organs Transplantation, Toulouse Rangueil University Hospital, Toulouse, France.,INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Paul Sabatier University, Toulouse, France
| | - Sebastien Lhomme
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Paul Sabatier University, Toulouse, France.,Virology Laboratory, National Reference Center for hepatitis E virus, Toulouse Purpan University Hospital, Toulouse, France
| | - Manon Nayrac
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France
| | - Martine Dubois
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Virology Laboratory, National Reference Center for hepatitis E virus, Toulouse Purpan University Hospital, Toulouse, France
| | - Mélanie Pucelle
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Virology Laboratory, National Reference Center for hepatitis E virus, Toulouse Purpan University Hospital, Toulouse, France
| | - Mary Requena
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Virology Laboratory, National Reference Center for hepatitis E virus, Toulouse Purpan University Hospital, Toulouse, France
| | - Marion Migueres
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Virology Laboratory, National Reference Center for hepatitis E virus, Toulouse Purpan University Hospital, Toulouse, France
| | - Florence Abravanel
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Paul Sabatier University, Toulouse, France.,Virology Laboratory, National Reference Center for hepatitis E virus, Toulouse Purpan University Hospital, Toulouse, France
| | - Jean Marie Peron
- Paul Sabatier University, Toulouse, France.,Hepatology and Gastroenterology Department, Toulouse Rangueil University Hospital, Toulouse, France
| | - Nicolas Carrere
- Paul Sabatier University, Toulouse, France.,Digestive Surgery Department, Toulouse Rangueil University Hospital, Toulouse, France
| | - Bertrand Suc
- Paul Sabatier University, Toulouse, France.,Digestive Surgery Department, Toulouse Rangueil University Hospital, Toulouse, France
| | - Pierre Delobel
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Paul Sabatier University, Toulouse, France.,Department of Infectious and Tropical Diseases, Toulouse Purpan University Hospital, Toulouse, France
| | - Nassim Kamar
- Department of Nephrology and Organs Transplantation, Toulouse Rangueil University Hospital, Toulouse, France.,INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France.,Paul Sabatier University, Toulouse, France
| | - Jacques Izopet
- INSERM UMR1043, Center for Pathophysiology of Toulouse Purpan, Toulouse, France .,Paul Sabatier University, Toulouse, France.,Virology Laboratory, National Reference Center for hepatitis E virus, Toulouse Purpan University Hospital, Toulouse, France
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This review highlights current knowledge on the dichotomous role played by T helper 17 cells (Th17)-polarized CD4 T cells in maintaining mucosal immunity homeostasis versus fueling HIV/simian immunodeficiency virus (SIV) replication/persistence during antiretroviral therapy (ART), with a focus on molecular mechanisms underlying these processes. RECENT FINDING Th17 cells bridge innate and adaptive immunity against pathogens at mucosal barrier surfaces. Th17 cells are located at portal sites of HIV/SIV entry, express a unique transcriptional/metabolic status compatible with viral replication, and represent the first targets of infection. The paucity of Th17 cells during HIV/SIV infection is caused by infection itself, but also by an altered Th17 differentiation, survival, and trafficking into mucosal sites. This causes major alterations of mucosal barrier integrity, microbial translocation, and disease progression. Unless initiated during the early acute infection phases, ART fails to restore the frequency/functionality of mucosal Th17 cells. A fraction of Th17 cells is long-lived and carry HIV reservoir during ART. Recent studies identified Th17-specific host factors controlling HIV transcription, a step untargeted by current ART. SUMMARY The identification of molecular mechanisms contributing to HIV replication/persistence in mucosal Th17 cells paves the way toward the design of new Th17-specific therapeutic strategies aimed at improving mucosal immunity in HIV-infected individuals.
Collapse
|
16
|
Zhao X, Li Y, Wang X, Wu J, Yuan Y, Lv S, Ren J. Synergistic association of FOXP3+ tumor infiltrating lymphocytes with CCL20 expressions with poor prognosis of primary breast cancer: A retrospective cohort study. Medicine (Baltimore) 2019; 98:e18403. [PMID: 31852159 PMCID: PMC6922488 DOI: 10.1097/md.0000000000018403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Studies have shown that forkhead/winged helix transcription factor P3 (FOXP3) tumor infiltrating lymphocytes (TILs) are intimately associated with invasion and survival of many invasive tumors. The inflammatory chemokine ligand 20 (CCL20) and its receptor CCR6 were found to be associated with tumor prognosis in some studies. Although increases in FOXP3 TILs infiltration and CCL20 expression have been revealed in several malignancies, their correlation in human breast tumors is as yet unclear.Surgically resected samples from 156 patients with invasive breast cancer (BC) were assessed for the expression of FOXP3 and CCL20 by immunohistochemistry. Correlation between their expressions and the association with clinicopathological characteristics and patient's prognosis were studied. Forty pairs of fresh BC and their nontumor adjacent tissues (NATs) in BC were carried out by real-time quantitative PCR (qRT-PCR) to evaluate the correlation between FOXP3 and CCL20 mRNA expression.CCL20 and FOXP3 TILs mRNA expression in tumor tissue demonstrated a high correlation (rs = 0.359, P < .001) in this cohort of breast cancer patients. Both elevated CCL20 expression and FOXP3 TILs infiltration were significantly correlated with high histological grade, positive human epidermal growth factor receptor-2 (HER2), high Ki67 index, and axillary lymph node metastases. Tumors with concomitant high expressions of both markers had the worst prognosis. Multivariate analysis showed that these 2 markers were independent predictors of overall survival. The patients with axillary lymph node metastases with the concomitant CCL20 high expression and increased FOXP3 TILs infiltration had the worst overall survival (OS) (P < .001), In lymph node-negative breast cancer patients, the status of CCL20 and FOXP3 was not related to OS (P = .22).The results suggest that CCL20 and FOXP3 TILs may have synergistic effects, and their upregulated expressions may lead to immune evasion in breast cancer. Combinatorial immunotherapeutic approaches aiming at blocking CCL20 and depleting FOXP3 might improve therapeutic efficacy in breast cancer patients.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
- Department of Surgical Breast Cancer, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yanping Li
- Department of Surgical Breast Cancer, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
| | - Jiangping Wu
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
| | - Yanhua Yuan
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
| | - Shuzhen Lv
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
- Department of Surgical Breast Cancer, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jun Ren
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
- Department of Surgery, Duke University Medical Center, Durham, NC, US
| |
Collapse
|
17
|
Differential Dynamics of Regulatory T-Cell and Th17 Cell Balance in Mesenteric Lymph Nodes and Blood following Early Antiretroviral Initiation during Acute Simian Immunodeficiency Virus Infection. J Virol 2019; 93:JVI.00371-19. [PMID: 31315987 PMCID: PMC6744245 DOI: 10.1128/jvi.00371-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022] Open
Abstract
Tregs contribute to SIV/HIV disease progression by inhibition of antiviral specific responses and effector T-cell proliferation. Tregs also cause tissue fibrosis via transforming growth factor β1 production and collagen deposition, which are associated with microbial translocation and generalized immune activation. Early ARV initiation upon viral exposure is recommended globally and results in improved immune function recovery and reduced viral persistence. Here, using an acute SIV infection model of rhesus macaques, we demonstrated for the first time that despite clear improvements in mucosal CD4 T cells, in contrast to blood, Treg frequencies in MLNs remained elevated following early ARV initiation. The particular Th17/Treg balance observed in MLNs can contribute, in part, to the maintenance of mucosal fibrosis during suppressive ARV treatment. Our results provide a better understanding of gut mucosal immune dynamics following early ARV initiation. These findings suggest that Treg-based treatments could serve as a novel immunotherapeutic approach to decrease gut mucosal damage during SIV/HIV infections. Increased frequencies of immunosuppressive regulatory T cells (Tregs) are associated with gut lymphoid tissue fibrosis and dysfunction which, in turn, contribute to disease progression in chronic simian immunodeficiency virus/human immunodeficiency virus (SIV/HIV) infection. Mesenteric lymph nodes (MLNs), which drain the large and small intestine, are critical sites for the induction and maintenance of gut mucosal immunity. However, the dynamics of Tregs in MLNs are not well understood due to the lack of accessibility to these tissues in HIV-infected individuals. Here, the dynamics of Tregs in blood and MLNs were assessed in SIV-infected rhesus macaques (RMs) following early antiretroviral drug (ARV) initiation. Early ARV initiation reduced T-cell immune activation, as assessed by HLA-DR/CD39 expression, and prevented the depletion of memory CCR6+ Th17 cells in both blood and MLNs. Untreated animals showed higher frequencies of Tregs, CD39+ Tregs, thymic Tregs, and new memory CD4 populations sharing similarity with Tregs as CTLA4+ PD1– and CTLA4+ PD1– FoxP3+ T cells. Despite early ARV treatment, the frequencies of these Treg subsets remained unchanged within the MLNs and, in contrast to blood normalization, the Th17/Treg ratio remained distorted in MLNs. Furthermore, our results highlighted that the expressions of IDO-1, TGFβ1 and collagen-1 mRNA remained unchanged in MLN of ARV-treated RMs. ARV interruption did not affect T-cell immune activation and Th17/Treg ratios in MLN. Altogether, our data demonstrated that early ARV initiation within the first few days of SIV infection is unable to reduce the frequencies and homing of various subsets of Tregs within the MLNs which, in turn, may result in tissue fibrosis, impairment in MLN function, and HIV persistence. IMPORTANCE Tregs contribute to SIV/HIV disease progression by inhibition of antiviral specific responses and effector T-cell proliferation. Tregs also cause tissue fibrosis via transforming growth factor β1 production and collagen deposition, which are associated with microbial translocation and generalized immune activation. Early ARV initiation upon viral exposure is recommended globally and results in improved immune function recovery and reduced viral persistence. Here, using an acute SIV infection model of rhesus macaques, we demonstrated for the first time that despite clear improvements in mucosal CD4 T cells, in contrast to blood, Treg frequencies in MLNs remained elevated following early ARV initiation. The particular Th17/Treg balance observed in MLNs can contribute, in part, to the maintenance of mucosal fibrosis during suppressive ARV treatment. Our results provide a better understanding of gut mucosal immune dynamics following early ARV initiation. These findings suggest that Treg-based treatments could serve as a novel immunotherapeutic approach to decrease gut mucosal damage during SIV/HIV infections.
Collapse
|
18
|
Taborda NA, Correa LA, Feria MG, Rugeles MT. The Spontaneous Control of HIV Replication is Characterized by Decreased Pathological Changes in the Gut-associated Lymphoid Tissue. Curr HIV Res 2019; 16:338-344. [PMID: 30706820 PMCID: PMC6446516 DOI: 10.2174/1570162x17666190130115113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/22/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND HIV infection induces alterations in the gut-associated lymphoid tissue (GALT) that constitutes the most important site for viral replication due to the extensive presence of effector memory T-cells. In the case of HIV-controllers, several studies have reported fewer peripheral alterations and conserved immune responses that correlate with viral control; however, the histopathological characterization of GALT in those patients is still missing. In this study, we evaluated pathological alterations in GALT, trying to associate them with clinical parameters of HIV infected patients with or without evidence of viral control. METHODS This study included eight HIV-controllers (antiretroviral treatment-naïve patients, with viral loads below 2.000 copies/mL for at least 1 year); 14 Noncontrollers (antiretroviral treatmentnaïve patients, with viral loads > 2.000 copies/mL and CD4+ T cells count > 250 cells/μL), and 12 uninfected donors. Biopsy fragments were obtained by rectosigmoidoscopy and stained with hematoxylin and eosin, silver methenamine, Ziehl Neelsen, and modified Ziehl Neelsen. RESULTS Histopathological findings in HIV-controllers were similar to those observed in the uninfected group. In contrast, noncontrollers exhibited several alterations including condyloma acuminate, squamous metaplasia and acute colitis. These alterations were associated with disease progression. CONCLUSION HIV-controllers exhibit lower pathological alterations in the gut tissue, associated with higher CD4 T cell count, and lower viral load.
Collapse
Affiliation(s)
- Natalia A Taborda
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia.,Grupo de Investigaciones Biomedicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporacion Universitaria Remington, Medellin, Colombia
| | - Luis A Correa
- Seccion de Dermatologia, Departamento de Medicina Interna, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia.,Laboratorio de Patología, Laboratorio Clínico VID, Obra de la Congregación Mariana, Medellín, Colombia
| | - Manuel Geronimo Feria
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| | - María T Rugeles
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| |
Collapse
|
19
|
Abstract
As our understanding of mucosal immunity increases, it is becoming clear that the host response to HIV-1 is more complex and nuanced than originally believed. The mucosal landscape is populated with a variety of specialized cell types whose functions include combating infectious agents while preserving commensal microbiota, maintaining barrier integrity, and ensuring immune homeostasis. Advances in multiparameter flow cytometry, gene expression analysis and bioinformatics have allowed more detailed characterization of these cell types and their roles in host defense than was previously possible. This review provides an overview of existing literature on immunity to HIV-1 and SIVmac in mucosal tissues of the female reproductive tract and the gastrointestinal tract, focusing on major effector cell populations and briefly summarizing new information on tissue resident memory T cells, Treg, Th17, Th22 and innate lymphocytes (ILC), subsets that have been studied primarily in the gastrointestinal mucosa.
Collapse
Affiliation(s)
- Barbara L Shacklett
- Department of Medical Microbiology and Immunology.,Division of Infectious Diseases, Department of Medicine, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
20
|
Loiseau C, Requena M, Nayrac M, Mavigner M, Cazabat M, Iscache AL, Carrere N, Suc B, Alric L, Izopet J, Delobel P. Increased CXCR3+ T Cells Impairs Recruitment of T-Helper Type 17 Cells via Interferon γ and Interleukin 18 in the Small Intestine Mucosa During Treated HIV-1 Infection. J Infect Dis 2019; 220:830-840. [DOI: 10.1093/infdis/jiz123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
Abstract
The restoration of CD4+ T cells, especially T-helper type 17 (Th17) cells, remains incomplete in the gut mucosa of most human immunodeficiency virus type 1 (HIV-1)–infected individuals despite sustained antiretroviral therapy (ART). Herein, we report an increase in the absolute number of CXCR3+ T cells in the duodenal mucosa during ART. The frequencies of Th1 and CXCR3+ CD8+ T cells were increased and negatively correlated with CCL20 and CCL25 expression in the mucosa. In ex vivo analyses, we showed that interferon γ, the main cytokine produced by Th1 and effector CD8+ T cells, downregulates the expression of CCL20 and CCL25 by small intestine enterocytes, while it increases the expression of CXCL9/10/11, the ligands of CXCR3. Interleukin 18, a pro-Th1 cytokine produced by enterocytes, also contributes to the downregulation of CCL20 expression and increases interferon γ production by Th1 cells. This could perpetuate an amplification loop for CXCR3-driven Th1 and effector CD8+ T cells recruitment to the gut, while impairing Th17 cells homing through the CCR6-CCL20 axis in treated HIV-1–infected individuals.
Collapse
Affiliation(s)
- C Loiseau
- INSERM, UMR1043, Toulouse, France
- aPresent affiliation: Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - M Requena
- Laboratoire de Virologie, Toulouse, France
| | - M Nayrac
- INSERM, UMR1043, Toulouse, France
| | - M Mavigner
- Department of Pediatrics, Atlanta, Georgia
- Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia
| | - M Cazabat
- Laboratoire de Virologie, Toulouse, France
| | | | - N Carrere
- Service de Chirurgie générale et digestive, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - B Suc
- Service de Chirurgie générale et digestive, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - L Alric
- Service de Médecine Interne, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
- IRD UMR152, Toulouse, France
| | - J Izopet
- INSERM, UMR1043, Toulouse, France
- Laboratoire de Virologie, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - P Delobel
- INSERM, UMR1043, Toulouse, France
- Service des Maladies Infectieuses et Tropicales, CHU de Toulouse, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
21
|
Basilissi M, Tincati C, Merlini E, Ancona G, Borghi E, Borgo F, Barassi A, d’Arminio Monforte A, Marchetti G. Mucosal cell populations may contribute to peripheral immune abnormalities in HIV-infected subjects introducing cART with moderate immune-suppression. PLoS One 2019; 14:e0212075. [PMID: 30763359 PMCID: PMC6375585 DOI: 10.1371/journal.pone.0212075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 01/25/2019] [Indexed: 12/13/2022] Open
Abstract
HIV infection causes the progressive depletion of CD4+ T-lymphocytes and profound modifications of T-cell homeostasis, which persist despite virologically-suppressive treatment and have been linked to a worse clinical outcome. Enduring alterations of the gastrointestinal tract may represent the underlying pathogenic mechanisms of these phenomena. Twenty-six HIV-infected subjects were assessed over a 12-month period following the introduction of antiretroviral therapy. 18 uninfected individuals were enrolled as controls. Parameters of peripheral T-cell homeostasis (activation, maturation), gastrointestinal function (microbial translocation, gut inflammation, fecal microbiota composition) and mucosal immunity (CD4+CCR6+CD161+, CD4+CCR9+α4β7+, stem cell memory CD4+/CD8+ T-cells) were assessed. CD4+CCR6+CD161+ cells were depleted in HIV-infected untreated subjects and maintained significantly lower levels compared to controls, despite the introduction of effective antiviral treatment. The frequency of gut-homing CD4+CCR9+α4β7+ cells was also impaired in untreated infection and correlated with the HIV RNA load and CD4+HLADR+CD38+; during therapy, we observed a contraction of this pool in the peripheral blood and the loss of its correlation with antigenic exposure/immune activation. A partial correction of the balance between stem cell memory pools and T-cell homeostasis was registered following treatment. In HIV-infected subjects with moderate immune-suppression, antiretroviral therapy has a marginal impact on mucosal immune populations which feature distinctive kinetics in the periphery, possibly reflecting their diverse recruitment from the blood to the mucosa. The persistent defects in mucosal immunity may fuel peripheral T-cell abnormalities through diverse mechanisms, including the production of IL-17/IL-22, cellular permissiveness to infection and regulation of T-lymphocyte maturation.
Collapse
Affiliation(s)
- Matteo Basilissi
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Italy
| | - Camilla Tincati
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Italy
- * E-mail:
| | - Esther Merlini
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Italy
| | - Giuseppe Ancona
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Italy
| | - Elisa Borghi
- Department of Health Sciences, Microbiology Laboratory, University of Milan, Italy
| | - Francesca Borgo
- Department of Health Sciences, Microbiology Laboratory, University of Milan, Italy
| | - Alessandra Barassi
- Department of Health Sciences, Biochemistry Laboratory, ASST Santi Paolo e Carlo, University of Milan, Italy
| | - Antonella d’Arminio Monforte
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Italy
| | - Giulia Marchetti
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Italy
| |
Collapse
|
22
|
Caruso MP, Falivene J, Holgado MP, Zurita DH, Laufer N, Castro C, Nico Á, Maeto C, Salido J, Pérez H, Salomón H, Cahn P, Sued O, Fink V, Turk G, Gherardi MM. Impact of HIV-ART on the restoration of Th17 and Treg cells in blood and female genital mucosa. Sci Rep 2019; 9:1978. [PMID: 30760809 PMCID: PMC6374372 DOI: 10.1038/s41598-019-38547-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 01/02/2019] [Indexed: 12/28/2022] Open
Abstract
The aim of this study was to evaluate the effectiveness of antiretroviral treatment (ART) on the proportion and functions of Th17 and Treg cells in peripheral blood and female genital tract (FGT) respectively. To this aim, samples from 41 HIV-neg, 33 HIV+ ART-naïve and 32 HIV+ ART+ subjects were obtained. In peripheral blood, altered Th17 and Th17/Treg proportions were normalized in HIV+ ART+, but certain abnormal Treg and activated T-cell proportions were still observed. In FGT, abnormal patterns of secretion for Th17-related cytokines were observed in cervical mononuclear cells (CMCs) from HIV+ women, even in those from HIV+ ART+, compared to the HIV-neg group. Moreover, these altered patterns of secretion were associated with diminished levels of CXCL5 and CXCL1 chemokines and with an immunoregulatory skew in the CCL17/CCL20 ratio in ectocervix samples of these women. Finally, ART did not restore proportions of Th17-precursor cells with gut-homing potential in PBMCs, and positive correlations between these cells and the levels of IL-17F and IL-21 production by CMCs may suggest that a better homing of these cells to the intestine could also imply a better restoration of these cells in the female genital tract. These results indicate that antiretroviral treatment did not restore Th17-related immune functions completely at the female mucosal level.
Collapse
Affiliation(s)
- María Paula Caruso
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Juliana Falivene
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - María Pía Holgado
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | | | - Natalia Laufer
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
- Hospital J.A. Fernández, Buenos Aires, Argentina
| | | | - Ángeles Nico
- Hospital J.A. Fernández, Buenos Aires, Argentina
| | - Cynthia Maeto
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Jimena Salido
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Héctor Pérez
- Hospital J.A. Fernández, Buenos Aires, Argentina
| | - Horacio Salomón
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Pedro Cahn
- Hospital J.A. Fernández, Buenos Aires, Argentina
- Fundación Huésped, Buenos Aires, Argentina
| | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina
| | - Valeria Fink
- Hospital J.A. Fernández, Buenos Aires, Argentina
- Fundación Huésped, Buenos Aires, Argentina
| | - Gabriela Turk
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - María Magdalena Gherardi
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
23
|
Cecchinato V, Uguccioni M. Insight on the regulation of chemokine activities. J Leukoc Biol 2018; 104:295-300. [PMID: 29668065 DOI: 10.1002/jlb.3mr0118-014r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 01/06/2023] Open
Abstract
The activity of chemokines is regulated by several mechanisms that control the final cellular response. The present review discusses the complexity of the regulation of the chemokine system, and the novel findings describing how in persistent infections, the expression of chemokine receptors on the surface of T cells does not correlate with their homing potential. Thanks to the latest advances in our comprehension of the chemokine system, novel approaches targeting chemokines, chemokine receptors, or protein of their signaling pathway should be considered in order to achieve a personalized therapy.
Collapse
Affiliation(s)
- Valentina Cecchinato
- Laboratory of "Chemokines in Immunity", Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Mariagrazia Uguccioni
- Laboratory of "Chemokines in Immunity", Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
24
|
Sun H, Geng W, Cui H, Liang G, Fu Y, Zhang Z, Jiang Y, Ding H, Xu J, Shang H. Alteration of CCR6 +CD95 +CD4 + naïve T cells in HIV-1 infected patients: Implication for clinical practice. Cell Immunol 2018; 327:47-53. [PMID: 29454646 DOI: 10.1016/j.cellimm.2018.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/09/2018] [Indexed: 01/10/2023]
Abstract
The profound deficiency of Th17 cells contributes to HIV disease progression. The mechanisms of their perturbation remain unclear. Recently, CCR6+CD95+CD4+ naïve T cells (CCR6+CD95+CD4+ TNA), identified as pre-committed Th17 precursors, were recognized as a subpopulation of CD4+ T cells with stem cell properties. Following phenotypical identification, we evaluated their level in patients during chronic HIV infection and following antiretroviral therapy (ART) using flow cytometry. The levels of CCR6+CD95+CD4+ TNA were decreased during chronic HIV infection and correlated with CD4+ T cell counts. Immunological responders harbored higher frequency of CCR6+CD95+CD4+ TNA, which was associated with CD4/CD8 T cell ratio. Immunological non-responders with lower frequency of CCR6+CD95+CD4+ TNA failed to exhibit a correlation between CCR6+CD95+CD4+ TNA and CCR6+CD95+CD4+ TCM, and displayed elevated ratio of CCR6+CD95+CD4+ TCM/TNA. The number of CCR6+CD95+CD4+ TNA was increased following early ART. These findings shed light on the importance of targeting pre-committed Th17 precursors that enhance immune reconstitution.
Collapse
Affiliation(s)
- Hong Sun
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Wenqing Geng
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Hualu Cui
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Guoxin Liang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Yajing Fu
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Zining Zhang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Yongjun Jiang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Haibo Ding
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Junjie Xu
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Hong Shang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China.
| |
Collapse
|
25
|
The Th17 Lineage: From Barrier Surfaces Homeostasis to Autoimmunity, Cancer, and HIV-1 Pathogenesis. Viruses 2017; 9:v9100303. [PMID: 29048384 PMCID: PMC5691654 DOI: 10.3390/v9100303] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022] Open
Abstract
The T helper 17 (Th17) cells represent a subset of CD4+ T-cells with unique effector functions, developmental plasticity, and stem-cell features. Th17 cells bridge innate and adaptive immunity against fungal and bacterial infections at skin and mucosal barrier surfaces. Although Th17 cells have been extensively studied in the context of autoimmunity, their role in various other pathologies is underexplored and remains an area of open investigation. This review summarizes the history of Th17 cell discovery and the current knowledge relative to the beneficial role of Th17 cells in maintaining mucosal immunity homeostasis. We further discuss the concept of Th17 pathogenicity in the context of autoimmunity, cancer, and HIV infection, and we review the most recent discoveries on molecular mechanisms regulating HIV replication/persistence in pathogenic Th17 cells. Finally, we stress the need for novel fundamental research discovery-based Th17-specific therapeutic interventions to treat pathogenic conditions associated with Th17 abnormalities, including HIV infection.
Collapse
|
26
|
Planas D, Zhang Y, Monteiro P, Goulet JP, Gosselin A, Grandvaux N, Hope TJ, Fassati A, Routy JP, Ancuta P. HIV-1 selectively targets gut-homing CCR6+CD4+ T cells via mTOR-dependent mechanisms. JCI Insight 2017; 2:93230. [PMID: 28768913 PMCID: PMC5543920 DOI: 10.1172/jci.insight.93230] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/29/2017] [Indexed: 12/30/2022] Open
Abstract
Gut-associated lymphoid tissues are enriched in CCR6+ Th17-polarized CD4+ T cells that contribute to HIV-1 persistence during antiretroviral therapy (ART). This raises the need for Th17-targeted immunotherapies. In an effort to identify mechanisms governing HIV-1 permissiveness/persistence in gut-homing Th17 cells, we analyzed the transcriptome of CCR6+ versus CCR6- T cells exposed to the gut-homing inducer retinoic acid (RA) and performed functional validations in colon biopsies of HIV-infected individuals receiving ART (HIV+ART). Although both CCR6+ and CCR6- T cells acquired gut-homing markers upon RA exposure, the modulation of unique sets of genes coincided with preferential HIV-1 replication in RA-treated CCR6+ T cells. This molecular signature included the upregulation of HIV-dependency factors acting at entry/postentry levels, such as the CCR5 and PI3K/Akt/mTORC1 signaling pathways. Of note, mTOR expression/phosphorylation was distinctively induced by RA in CCR6+ T cells. Consistently, mTOR inhibitors counteracted the effect of RA on HIV replication in vitro and viral reactivation in CD4+ T cells from HIV+ART individuals via postentry mechanisms independent of CCR5. Finally, CCR6+ versus CCR6- T cells infiltrating the colons of HIV+ART individuals expressed unique molecular signatures, including higher levels of CCR5, integrin β7, and mTOR phosphorylation. Together, our results identify mTOR as a druggable key regulator of HIV permissiveness in gut-homing CCR6+ T cells.
Collapse
Affiliation(s)
- Delphine Planas
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Québec, Canada
- Département of microbiologie, infectiologie et immunologie, Université de Montréal, Faculté de Médecine, Montreal, Québec, Canada
| | - Yuwei Zhang
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Québec, Canada
- Département of microbiologie, infectiologie et immunologie, Université de Montréal, Faculté de Médecine, Montreal, Québec, Canada
| | - Patricia Monteiro
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Québec, Canada
- Département of microbiologie, infectiologie et immunologie, Université de Montréal, Faculté de Médecine, Montreal, Québec, Canada
| | | | - Annie Gosselin
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Québec, Canada
| | - Nathalie Grandvaux
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Québec, Canada
- Faculté de Médecine, Département of biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
| | - Thomas J. Hope
- Department of Cellular and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Jean-Pierre Routy
- Chronic Viral Illness Service and Research Institute and
- Division of Hematology, McGill University Health Centre, Montreal, Québec, Canada
| | - Petronela Ancuta
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Québec, Canada
- Département of microbiologie, infectiologie et immunologie, Université de Montréal, Faculté de Médecine, Montreal, Québec, Canada
| |
Collapse
|
27
|
McGary CS, Alvarez X, Harrington S, Cervasi B, Ryan ES, Iriele RI, Paganini S, Harper J, Easley K, Silvestri G, Ansari AA, Lichterfeld M, Micci L, Paiardini M. The loss of CCR6 + and CD161 + CD4 + T-cell homeostasis contributes to disease progression in SIV-infected rhesus macaques. Mucosal Immunol 2017; 10:1082-1096. [PMID: 28051083 PMCID: PMC5474141 DOI: 10.1038/mi.2016.116] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/10/2016] [Indexed: 02/04/2023]
Abstract
Although previous studies have shown that CD4+ T cells expressing CCR6 and CD161 are depleted from blood during HIV infection, the mechanisms underlying their loss remain unclear. In this study, we investigated how the homeostasis of CCR6+ and CD161+ CD4+ T cells contributes to SIV disease progression and the mechanisms responsible for their loss from circulation. By comparing SIV infection in rhesus macaques (RMs) and natural host sooty mangabeys (SMs), we found that the loss of CCR6+ and CD161+ CD4+ T cells from circulation is a distinguishing feature of progressive SIV infection in RMs. Furthermore, while viral infection critically contributes to the loss of CD161+CCR6-CD4+ T cells, a redistribution of CCR6+CD161- and CCR6+CD161+CD4+ T cells from the blood to the rectal mucosa is a chief mechanism for their loss during SIV infection. Finally, we provide evidence that the accumulation of CCR6+CD4+ T cells in the mucosa is damaging to the host by demonstrating their reduction from this site following initiation of antiretroviral therapy in SIV-infected RMs and their lack of accumulation in SIV-infected SMs. These data emphasize the importance of maintaining CCR6+ and CD161+ CD4+ T-cell homeostasis, particularly in the mucosa, to prevent disease progression during pathogenic HIV/SIV infection.
Collapse
Affiliation(s)
- Colleen S. McGary
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Xavier Alvarez
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA
| | - Sean Harrington
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, MA
| | - Barbara Cervasi
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Emily S. Ryan
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Robin I. Iriele
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Sara Paganini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Justin Harper
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Kirk Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Aftab A. Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Mathias Lichterfeld
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, MA
| | - Luca Micci
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
28
|
Diversity and functions of intestinal mononuclear phagocytes. Mucosal Immunol 2017; 10:845-864. [PMID: 28378807 DOI: 10.1038/mi.2017.22] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/16/2017] [Accepted: 02/22/2017] [Indexed: 02/04/2023]
Abstract
The intestinal lamina propria (LP) contains a diverse array of mononuclear phagocyte (MNP) subsets, including conventional dendritic cells (cDC), monocytes and tissue-resident macrophages (mφ) that collectively play an essential role in mucosal homeostasis, infection and inflammation. In the current review we discuss the function of intestinal cDC and monocyte-derived MNP, highlighting how these subsets play several non-redundant roles in the regulation of intestinal immune responses. While much remains to be learnt, recent findings also underline how the various populations of MNP adapt to deal with the challenges specific to their environment. Understanding these processes should help target individual subsets for 'fine tuning' immunological responses within the intestine, a process that may be of relevance both for the treatment of inflammatory bowel disease (IBD) and for optimized vaccine design.
Collapse
|
29
|
Lee JJ, Kao KC, Chiu YL, Jung CJ, Liu CJ, Cheng SJ, Chang YL, Ko JY, Chia JS. Enrichment of Human CCR6 + Regulatory T Cells with Superior Suppressive Activity in Oral Cancer. THE JOURNAL OF IMMUNOLOGY 2017; 199:467-476. [PMID: 28600287 DOI: 10.4049/jimmunol.1601815] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 05/12/2017] [Indexed: 01/29/2023]
Abstract
Human oral squamous cell carcinoma (OSCC) constitutes an inflammatory microenvironment enriched with chemokines such as CCL20, which promote cancer cell invasion and tumor progression. We found that in OSCC there is a correlation between the expression of CCL20 and FOXP3 mRNA. Therefore, we hypothesized that OSCC may favor the recruitment and retention of regulatory T (Treg) cells that express the CCL20 receptor, CCR6. Interestingly, most (∼60%) peripheral blood Treg cells express CCR6, and CCR6+ Treg cells exhibit an activated effector/memory phenotype. In contrast, a significant portion (>30%) of CCR6- Treg cells were found to be CD45RA+ naive Treg cells. Compared to CCR6- naive or memory Treg cells, CCR6+ Treg cells exhibit stronger suppressive activity and display higher FOXP3 expression along with lower methylation at the Treg-specific demethylated region of the FOXP3 gene. This predominance of CCR6+ Treg cells was also found in the draining lymph nodes and tumor-infiltrating lymphocytes of OSCC patients with early or late clinical staging. Moreover, CCR6+ Treg cells isolated from tumor-infiltrating lymphocytes or draining lymph nodes maintained similar phenotypic and suppressive characteristics ex vivo as did their counterparts isolated from peripheral blood. These results suggest that CCR6 marks activated effector or memory Treg phenotypes with superior suppressive activity in humans.
Collapse
Affiliation(s)
- Jang-Jaer Lee
- Department of Oral Maxillofacial Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Kung-Chi Kao
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yen-Ling Chiu
- Department of Nephrology, Far Eastern Memorial Hospital, Taipei 220, Taiwan.,Graduate Program of Biomedical Informatics, Yuan Ze University, Taoyuan 320, Taiwan
| | - Chiau-Jing Jung
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chung-Ji Liu
- Department of Oral Maxillofacial Surgery, Taipei MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Shih-Jung Cheng
- Department of Oral Maxillofacial Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Yen-Liang Chang
- Department of Otolaryngology, Fu Jen Catholic University College of Medicine, New Taipei City 24205, Taiwan; and
| | - Jenq-Yuh Ko
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Jean-San Chia
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| |
Collapse
|
30
|
Gu T, De Jesus M, Gallagher HC, Burris TP, Egilmez NK. Oral IL-10 suppresses colon carcinogenesis via elimination of pathogenicCD4 + T-cells and induction of antitumor CD8 + T-cell activity. Oncoimmunology 2017; 6:e1319027. [PMID: 28680752 DOI: 10.1080/2162402x.2017.1319027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/07/2017] [Accepted: 04/07/2017] [Indexed: 12/19/2022] Open
Abstract
An oral sustained-release formulation of Interleukin-10 suppressed tumor growth and enhanced survival in the APCmin/+/Bacteroides fragilis spontaneous colon cancer model. Therapeutic benefit was associated with a 5-fold reduction in CD4+RORγt+Foxp3-IL-17+ T-helper cell, CD4+RORγt+Foxp3+IL-17+ pathogenic T-regulatory cell and CD4+RORγt-Foxp3+IL-17- conventional T-regulatory cell numbers and a concurrent 2-fold enhancement in CD8+ T-cell activity in the colon. Selective subset depletion and functional blockade studies demonstrated that at steady-state CD4+RORγt+IL-17+ T-cell subsets and CD4+Foxp3+ cTreg supported tumorigenesis, whereas CD8+ cytotoxic T-lymphocytes impeded tumor progression following IL-10 therapy. Suppression of tumor growth by CD8+ T-cells was associated with enhanced tumor infiltration and cytotoxic granule exocytosis. These findings establish the utility of oral IL-10 as a potential new therapeutic in the management of colon cancer and shed light on the cellular mechanisms that underlie its antitumor activity.
Collapse
Affiliation(s)
- Tao Gu
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Magdia De Jesus
- Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY, USA.,Wadsworth Center, New York State Department of Health, David Axelrod Institute, Albany, NY, USA
| | - Heather C Gallagher
- Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY, USA.,Wadsworth Center, New York State Department of Health, David Axelrod Institute, Albany, NY, USA
| | - Thomas P Burris
- Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
31
|
Lee AYS, Körner H. CCR6/CCL20 chemokine axis in human immunodeficiency virus immunity and pathogenesis. J Gen Virol 2017; 98:338-344. [PMID: 28005525 DOI: 10.1099/jgv.0.000691] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent studies in human immunodeficiency virus (HIV) have garnered interest for the role of CC chemokine receptor 6 (CCR6) and its known ligands, CC chemokine ligand 20 (CCL20) and human β-defensins, in viral entry, dissemination and antiviral immunity. Several studies have suggested that CCR6 may also act as a weak co-receptor of HIV entry, in addition to the canonical CXC chemokine receptor 4 (CXCR4) and CCR5. However, the pathogenic significance has yet to be demonstrated as the observations for preferential infection of CD4+CCR6+ over CD4+CCR6- T cells appear to be independent of CCR6 expression. This indicates means for preferential infection other than CCR6 co-receptor use. Attention has also turned to the inadvertent role of the CCR6/CCL20 axis in attracting key immune cells, including TH17 cells and dendritic cells, to sites of infection and propagating the virus to other sites of the body. This review article will summarize the latest evidence that the CCR6/CCL20 chemokine axis is playing an important role in HIV pathogenesis and immunity. Further work with in vivo studies is needed to establish the biological and, hence, therapeutic significance of these findings.
Collapse
Affiliation(s)
- Adrian Y S Lee
- Western Health, Melbourne, Victoria, Australia
- School of Medicine, University of Tasmania, Tasmania, Australia
| | - Heinrich Körner
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Engineering Technology Research Center of Anti-inflammatory and Immunodrugs in Anhui Province, Hefei, Anhui Province, PR China
| |
Collapse
|
32
|
Scharschmidt TC, Vasquez KS, Pauli ML, Leitner EG, Chu K, Truong HA, Lowe MM, Sanchez Rodriguez R, Ali N, Laszik ZG, Sonnenburg JL, Millar SE, Rosenblum MD. Commensal Microbes and Hair Follicle Morphogenesis Coordinately Drive Treg Migration into Neonatal Skin. Cell Host Microbe 2017; 21:467-477.e5. [PMID: 28343820 DOI: 10.1016/j.chom.2017.03.001] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/13/2017] [Accepted: 03/01/2017] [Indexed: 01/02/2023]
Abstract
Regulatory T cells (Tregs) are required to establish immune tolerance to commensal microbes. Tregs accumulate abruptly in the skin during a defined window of postnatal tissue development. However, the mechanisms mediating Treg migration to neonatal skin are unknown. Here we show that hair follicle (HF) development facilitates the accumulation of Tregs in neonatal skin and that upon skin entry these cells localize to HFs, a primary reservoir for skin commensals. Further, germ-free neonates had reduced skin Tregs indicating that commensal microbes augment Treg accumulation. We identified Ccl20 as a HF-derived, microbiota-dependent chemokine and found its receptor, Ccr6, to be preferentially expressed by Tregs in neonatal skin. The Ccl20-Ccr6 pathway mediated Treg migration in vitro and in vivo. Thus, HF morphogenesis, commensal microbe colonization, and local chemokine production work in concert to recruit Tregs into neonatal skin, thereby establishing this tissue Treg niche early in life.
Collapse
Affiliation(s)
- Tiffany C Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Kimberly S Vasquez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Mariela L Pauli
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Elizabeth G Leitner
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Kevin Chu
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Hong-An Truong
- Immuno-Oncology Group, Bristol-Meyers Squibb, Redwood City, CA, 94063, USA
| | - Margaret M Lowe
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Robert Sanchez Rodriguez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Niwa Ali
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Zoltan G Laszik
- Department of Pathology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sarah E Millar
- Departments of Dermatology and Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
33
|
Toll-Like Receptor 2 Ligation Enhances HIV-1 Replication in Activated CCR6+ CD4+ T Cells by Increasing Virus Entry and Establishing a More Permissive Environment to Infection. J Virol 2017; 91:JVI.01402-16. [PMID: 27928019 DOI: 10.1128/jvi.01402-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/01/2016] [Indexed: 12/14/2022] Open
Abstract
In this study, we investigated the effect of Toll-like receptor 2 (TLR2) ligation on the permissiveness of activated CD4+ T cells to HIV-1 infection by focusing our experiments on the relative susceptibility of cell subsets based on their expression of CCR6. Purified primary human CD4+ T cells were first subjected to a CD3/CD28 costimulation before treatment with the TLR2 agonist Pam3CSK4. Finally, cells were inoculated with R5-tropic HIV-1 particles that permit us to study the effect of TLR2 triggering on virus production at both population and single-cell levels. We report here that HIV-1 replication is augmented in CD3/CD28-costimulated CCR6+ CD4+ T cells upon engagement of the cell surface TLR2. Additional studies indicate that a higher virus entry and polymerization of the cortical actin are seen in this cell subset following TLR2 stimulation. A TLR2-mediated increase in the level of phosphorylated NF-κB p65 subunit was also detected in CD3/CD28-costimulated CCR6+ CD4+ T cells. We propose that, upon antigenic presentation, an engagement of TLR2 acts specifically on CCR6+ CD4+ T cells by promoting virus entry in an intracellular milieu more favorable for productive HIV-1 infection. IMPORTANCE Following primary infection, HIV-1 induces an immunological and structural disruption of the gut mucosa, leading to bacterial translocation and release of microbial components in the bloodstream. These pathogen-derived constituents include several agonists of Toll-like receptors that may affect gut-homing CD4+ T cells, such as those expressing the chemokine receptor CCR6, which are highly permissive to HIV-1 infection. We demonstrate that TLR2 ligation in CD3/CD28-costimulated CCR6+ CD4+ T cells leads to enhanced virus production. Our results highlight the potential impact of bacterial translocation on the overall permissiveness of CCR6+ CD4+ T cells to productive HIV-1 infection.
Collapse
|
34
|
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: 119] [Impact Index Per Article: 14.9] [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
|