1
|
Karim F, Riou C, Bernstein M, Jule Z, Lustig G, van Graan S, Keeton RS, Upton JL, Ganga Y, Khan K, Reedoy K, Mazibuko M, Govender K, Thambu K, Ngcobo N, Venter E, Makhado Z, Hanekom W, von Gottberg A, Hoque M, Karim QA, Abdool Karim SS, Manickchund N, Magula N, Gosnell BI, Lessells RJ, Moore PL, Burgers WA, de Oliveira T, Moosa MYS, Sigal A. Clearance of persistent SARS-CoV-2 associates with increased neutralizing antibodies in advanced HIV disease post-ART initiation. Nat Commun 2024; 15:2360. [PMID: 38491050 PMCID: PMC10943233 DOI: 10.1038/s41467-024-46673-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
SARS-CoV-2 clearance requires adaptive immunity but the contribution of neutralizing antibodies and T cells in different immune states is unclear. Here we ask which adaptive immune responses associate with clearance of long-term SARS-CoV-2 infection in HIV-mediated immunosuppression after suppressive antiretroviral therapy (ART) initiation. We assembled a cohort of SARS-CoV-2 infected people in South Africa (n = 994) including participants with advanced HIV disease characterized by immunosuppression due to T cell depletion. Fifty-four percent of participants with advanced HIV disease had prolonged SARS-CoV-2 infection (>1 month). In the five vaccinated participants with advanced HIV disease tested, SARS-CoV-2 clearance associates with emergence of neutralizing antibodies but not SARS-CoV-2 specific CD8 T cells, while CD4 T cell responses were not determined due to low cell numbers. Further, complete HIV suppression is not required for clearance, although it is necessary for an effective vaccine response. Persistent SARS-CoV-2 infection led to SARS-CoV-2 evolution, including virus with extensive neutralization escape in a Delta variant infected participant. The results provide evidence that neutralizing antibodies are required for SARS-CoV-2 clearance in HIV-mediated immunosuppression recovery, and that suppressive ART is necessary to curtail evolution of co-infecting pathogens to reduce individual health consequences as well as public health risk linked with generation of escape mutants.
Collapse
Affiliation(s)
- Farina Karim
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | | | - Zesuliwe Jule
- Africa Health Research Institute, Durban, South Africa
| | - Gila Lustig
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Strauss van Graan
- SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Roanne S Keeton
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | | | - Yashica Ganga
- Africa Health Research Institute, Durban, South Africa
| | - Khadija Khan
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Kajal Reedoy
- Africa Health Research Institute, Durban, South Africa
| | | | | | | | | | - Elizabeth Venter
- SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Zanele Makhado
- SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Willem Hanekom
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, UK
| | - Anne von Gottberg
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Monjurul Hoque
- KwaDabeka Community Health Centre, KwaDabeka, South Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Nithendra Manickchund
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Nombulelo Magula
- Department of Internal Medicine, Nelson R. Mandela School of Medicine, University of Kwa-Zulu Natal, Durban, South Africa
| | - Bernadett I Gosnell
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Richard J Lessells
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform, Durban, South Africa
| | - Penny L Moore
- SAMRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Tulio de Oliveira
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform, Durban, South Africa
- Centre for Epidemic Response and Innovation, School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Mahomed-Yunus S Moosa
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa.
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa.
| |
Collapse
|
2
|
Ikeogu N, Ajibola O, Zayats R, Murooka TT. Identifying physiological tissue niches that support the HIV reservoir in T cells. mBio 2023; 14:e0205323. [PMID: 37747190 PMCID: PMC10653859 DOI: 10.1128/mbio.02053-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Successful antiretroviral therapy (ART) can efficiently suppress Human Immunodeficiency Virus-1 (HIV-1) replication to undetectable levels, but rare populations of infected memory CD4+ T cells continue to persist, complicating viral eradication efforts. Memory T cells utilize distinct homing and adhesion molecules to enter, exit, or establish residence at diverse tissue sites, integrating cellular and environmental cues that maintain homeostasis and life-long protection against pathogens. Critical roles for T cell receptor and cytokine signals driving clonal expansion and memory generation during immunity generation are well established, but whether HIV-infected T cells can utilize similar mechanisms for their own long-term survival is unclear. How infected, but transcriptionally silent T cells maintain their recirculation potential through blood and peripheral tissues, or whether they acquire new capabilities to establish unique peripheral tissue niches, is also not well understood. In this review, we will discuss the cellular and molecular cues that are important for memory T cell homeostasis and highlight opportunities for HIV to hijack normal immunological processes to establish long-term viral persistence.
Collapse
Affiliation(s)
- Nnamdi Ikeogu
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Oluwaseun Ajibola
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Romaniya Zayats
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Thomas T. Murooka
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
3
|
Allué-Guardia A, Torrelles JB, Sigal A. Tuberculosis and COVID-19 in the elderly: factors driving a higher burden of disease. Front Immunol 2023; 14:1250198. [PMID: 37841265 PMCID: PMC10569613 DOI: 10.3389/fimmu.2023.1250198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) and SARS-CoV-2 are both infections that can lead to severe disease in the lower lung. However, these two infections are caused by very different pathogens (Mycobacterium vs. virus), they have different mechanisms of pathogenesis and immune response, and differ in how long the infection lasts. Despite the differences, SARS-CoV-2 and M.tb share a common feature, which is also frequently observed in other respiratory infections: the burden of disease in the elderly is greater. Here, we discuss possible reasons for the higher burden in older adults, including the effect of co-morbidities, deterioration of the lung environment, auto-immunity, and a reduced antibody response. While the answer is likely to be multifactorial, understanding the main drivers across different infections may allow us to design broader interventions that increase the health-span of older people.
Collapse
Affiliation(s)
- Anna Allué-Guardia
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jordi B. Torrelles
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, United States
- International Center for the Advancement of Research and Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
4
|
Zinc Finger Protein BCL11A Contributes to the Abortive Infection of Hirame novirhabdovirus (HIRRV) in B Lymphocytes of Flounder (Paralichthys olivaceus). J Virol 2022; 96:e0147022. [PMID: 36448803 PMCID: PMC9769382 DOI: 10.1128/jvi.01470-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Hirame novirhabdovirus (HIRRV) infection is characterized by a pronounced viremia, and the high viral load is typically detected in immune-related organs and the circulatory system. In the present study, we demonstrated that HIRRV has the capacity to invade part of flounder membrane-bound IgM (mIgM+) B lymphocyte. Eight quantitative real-time PCR (qRT-PCR) standard curves involving HIRRV genomic RNA (gRNA), cRNA, and six mRNAs were established based on the strand-specific reverse transcription performed with tagged primers. It was revealed that viral RNA synthesis, especially the replication of gRNA, was inhibited in B cells, and the intracellular HIRRV even failed to produce infectious viral particles. Moreover, a range of genes with nucleic acid binding activity or related to viral infection were screened out based on the transcriptome analysis of HIRRV-infected B cells, and five molecules were further selected because of their different expression patterns in HIRRV-infected B cells and hirame natural embryo (HINAE) cells. The overexpression of these genes followed by HIRRV infection and RNA binding protein immunoprecipitation (RIP) assay revealed that the flounder B cell lymphoma/leukemia 11A (BCL11A), a highly conserved zinc finger transcription factor, is able to inhibit the proliferation of HIRRV by binding with full-length viral RNA mainly via its zinc finger domains at the C terminus. In conclusion, these data indicated that the high transcriptional activity of BCL11A in flounder mIgM+ B lymphocytes is a crucial factor for the abortive infection of HIRRV, and our findings provide new insights into the interaction between HIRRV and teleost B cells. IMPORTANCE HIRRV is a fish rhabdovirus that is considered as an important pathogen threatening the fish farming industry represented by flounder because of its high infectivity and fatality rate. To date, research toward understanding the complex pathogenic mechanism of HIRRV is still in its infancy and faces many challenges. Exploration of the relationship between HIRRV and its target cells is interesting and necessary. Here, we revealed that flounder mIgM+ B cells are capable of suppressing viral RNA synthesis and result in an unproductive infection of HIRRV. In addition, our results demonstrated that zinc finger protein BCL11A, a transcription factor in B cells, is able to suppress the replication of HIRRV. These findings increased our understanding of the underlying characteristics of HIRRV infection and revealed a novel antiviral mechanism against HIRRV based on the host restriction factor in teleost B cells, which sheds new light on the research into HIRRV control.
Collapse
|
5
|
Zhao J, Chen H, Wan Z, Yu T, Liu Q, Shui J, Wang H, Peng J, Tang S. Evaluation of antiretroviral therapy effect and prognosis between HIV-1 recent and long-term infection based on a rapid recent infection testing algorithm. Front Microbiol 2022; 13:1004960. [PMID: 36483196 PMCID: PMC9722761 DOI: 10.3389/fmicb.2022.1004960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/14/2022] [Indexed: 08/30/2023] Open
Abstract
Early diagnosis of HIV-1 infection and immediate initiation of combination antiretroviral therapy (cART) are important for achieving better virological suppression and quicker immune reconstitution. However, no serological HIV-1 recency testing assay has been approved for clinical use, and the real-world clinical outcomes remain to be explored for the subjects with HIV-1 recent infection (RI) or long-term infection (LI) when antiretroviral therapy is initiated. In this study, a HIV-1 rapid recent-infection testing strip (RRITS) was developed and incorporated into the recent infection testing algorithms (RITAs) to distinguish HIV-1 RI and LI and to assess their clinical outcomes including virological response, the recovery of CD4+ T-cell count and CD4/CD8 ratio and the probability of survival. We found that the concordance between our RRITS and the commercially available LAg-Avidity EIA was 97.13% and 90.63% when detecting the longitudinal and cross-sectional HIV-1 positive samples, respectively. Among the 200 HIV-1 patients analyzed, 22.5% (45/200) of them were RI patients and 77.5% (155/200) were chronically infected and 30% (60/200) of them were AIDS patients. After cART, 4.1% (5/155) of the LI patients showed virological rebound, but none in the RI group. The proportion of CD4+ T-cell count >500 cells/mm3 was significantly higher in RI patients than in LI after 2 years of cART with a hazard ratio (HR) of 2.6 (95% CI: 1.9, 3.6, p < 0.0001) while the probability of CD4/CD8 = 1 was higher in RI than in LI group with a HR of 3.6 (95% CI: 2.2, 5.7, p < 0.0001). Furthermore, the immunological recovery speed was 16 cells/mm3/month for CD4+ T-cell and 0.043/month for the ratio of CD4/CD8 in the RI group, and was bigger in the RI group than in the LI patients (p < 0.05) during the 1st year of cART. The survival probability for LI patients was significantly lower than that for RI patients (p < 0.001). Our results indicated that RRITS combined with RITAs could successfully distinguish HIV-1 RI and LI patients whose clinical outcomes were significantly different after cART. The rapid HIV-1 recency test provides a feasible assay for diagnosing HIV-1 recent infection and a useful tool for predicting the outcomes of HIV-1 patients.
Collapse
Affiliation(s)
- Jianhui Zhao
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hongjie Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhengwei Wan
- Department of Health Management and Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Yu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Quanxun Liu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jingwei Shui
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haiying Wang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jie Peng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shixing Tang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Lopez P, Ajibola O, Pagliuzza A, Zayats R, Koh WH, Herschhorn A, Chomont N, Murooka TT. T cell migration potentiates HIV infection by enhancing viral fusion and integration. Cell Rep 2022; 38:110406. [PMID: 35196491 DOI: 10.1016/j.celrep.2022.110406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 10/27/2021] [Accepted: 01/27/2022] [Indexed: 11/26/2022] Open
Abstract
T cells actively migrate along reticular networks within lymphoid organs in search for cognate antigen, but how these behaviors impact HIV entry and infection is unclear. Here, we show that migratory T cells in 3D collagen matrix display significantly enhanced infection and integration by cell-free R5-tropic lab adapted and transmitted/founder molecular HIV clones in the absence of exogenous cytokines or cationic polymers. Using two different collagen matrices that either support or restrict T cell migration, we observe high levels of HIV fusion in migratory T cells, whereas non-motile T cells display low viral entry and integration. Motile T cells were less sensitive to combination antiretroviral drugs and were able to freely migrate into regions with high HIV densities, resulting in high infection rates. Together, our studies indicate that the environmental context in which initial HIV-T cell encounters occur modulates HIV-1 entry and integration efficiencies.
Collapse
Affiliation(s)
- Paul Lopez
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Oluwaseun Ajibola
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Amelie Pagliuzza
- Department of Microbiology, Infectiology and Immunology, Centre de recherche du CHUM and Université de Montréal, Montreal, QC, Canada
| | - Romaniya Zayats
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Wan Hon Koh
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Alon Herschhorn
- Department of Medicine, Division of Infectious Diseases and International Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Nicolas Chomont
- Department of Microbiology, Infectiology and Immunology, Centre de recherche du CHUM and Université de Montréal, Montreal, QC, Canada
| | - Thomas T Murooka
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada; Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
7
|
Li X, Zhao J, Naini SM, Sabiu G, Tullius SG, Shin SR, Bromberg JS, Fiorina P, Tsokos GC, Abdi R, Kasinath V. Kidney-Draining Lymph Node Fibrosis Following Unilateral Ureteral Obstruction. Front Immunol 2021; 12:768412. [PMID: 35024041 PMCID: PMC8744208 DOI: 10.3389/fimmu.2021.768412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/01/2021] [Indexed: 12/02/2022] Open
Abstract
Although the primary organ has been the subject of intense investigation in the field of organ fibrosis over the past several decades, the presence of lymph node fibrosis due to persistent activation of the immune response in its partner organ remains largely unknown. Previously, we demonstrated that activation of the immune response following ischemia-reperfusion injury (IRI) and crescentic glomerulonephritis (CGN) in the kidney was associated with extracellular matrix (ECM) production by fibroblastic reticular cells (FRCs) of the kidney-draining lymph node (KLN). Here, we sought to determine whether FRCs in the KLN become similarly fibrogenic following unilateral ureteral obstruction (UUO) of the kidney. We subjected 6-8-week-old C57BL/6J mice to UUO for 2, 7, and 14 days. We examined the microarchitecture of the kidney and KLN by immunofluorescence staining at each timepoint, and we quantified immune cell populations in the KLN by flow cytometry. The contralateral kidney unaffected by UUO and its partner KLN were used as controls. We found through immunofluorescence staining that FRCs increased production of ECM fibers and remodeled the microarchitecture of the UUO KLN, contributing to fibrosis that mirrored the changes in the kidney. We also observed by flow cytometry that the populations of CD11b+ antigen-presenting cells, CD11c+ dendritic cells, and activated CD4+ and CD8+ T cells were significantly higher in the UUO KLN than the KLN draining the unaffected contralateral kidney. Expression of the TGFβ/TGFβR signaling pathway was upregulated and colocalized with FRCs in the UUO KLNs, suggesting a possible mechanism behind the fibrosis. Both release of ureteral ligation at 2 days following UUO and depletion of FRCs at the time of injury onset halted the progression of fibrosis in both the kidney and the KLN. These findings for the first time highlight the association between fibrosis both in the kidney and the KLN during UUO, and they lay the groundwork for future studies that will investigate more deeply the mechanisms behind the connection between FRCs and KLN fibrosis.
Collapse
Affiliation(s)
- Xiaofei Li
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Jing Zhao
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Said Movahedi Naini
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Gianmarco Sabiu
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Stefan G. Tullius
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, United States
| | - Jonathan S. Bromberg
- Departments of Surgery and Microbiology and Immunology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MA, United States
| | - Paolo Fiorina
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - George C. Tsokos
- Division of Rheumatology and Department of Immunology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Reza Abdi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Vivek Kasinath
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
Calvet-Mirabent M, Claiborne DT, Deruaz M, Tanno S, Serra C, Delgado-Arévalo C, Sánchez-Cerrillo I, de Los Santos I, Sanz J, García-Fraile L, Sánchez-Madrid F, Alfranca A, Muñoz-Fernández MÁ, Allen TM, Buzón MJ, Balazs A, Vrbanac V, Martín-Gayo E. Poly I:C and STING agonist-primed DC increase lymphoid tissue polyfunctional HIV-1-specific CD8 + T cells and limit CD4 + T cell loss in BLT mice. Eur J Immunol 2021; 52:447-461. [PMID: 34935145 DOI: 10.1002/eji.202149502] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/19/2021] [Accepted: 12/14/2021] [Indexed: 11/11/2022]
Abstract
Effective function of CD8+ T cells and enhanced innate activation of dendritic cells (DC) in response to HIV-1 is linked to protective antiviral immunity in controllers. Manipulation of DC targeting the master regulator TANK-binding Kinase 1 (TBK1) might be useful to acquire controller-like properties. Here, we evaluated the impact of the combination of 2´3´-c´diAM(PS)2 and Poly I:C as potential adjuvants capable of potentiating DC´s abilities to induce polyfunctional HIV-1 specific CD8+ T cell responses in vitro and in vivo using a humanized BLT mouse model. Adjuvant combination enhanced TBK-1 phosphorylation and IL-12 and IFNβ expression on DC and increased their ability to activate polyfunctional HIV-1-specific CD8+ T cells in vitro. Moreover, higher proportions of hBLT mice vaccinated with ADJ-DC exhibited less severe CD4+ T cell depletion following HIV-1 infection compared to control groups. This was associated with infiltration of CD8+ T cells in the white pulp from the spleen, reduced spread of infected p24+ cells to lymph node and with preserved abilities of CD8+ T cells from the spleen and blood of vaccinated animals to induce specific polyfunctional responses upon antigen stimulation. Therefore, priming of DC with Poly I:C and STING agonists might be useful for future HIV-1 vaccine studies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Marta Calvet-Mirabent
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| | | | - Maud Deruaz
- Human Immune System Mouse Program from Massachusetts General Hospital, Boston.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Serah Tanno
- Ragon Institute of MGH, MIT and Harvard.,Human Immune System Mouse Program from Massachusetts General Hospital, Boston
| | - Carla Serra
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona
| | - Cristina Delgado-Arévalo
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| | - Ildefonso Sánchez-Cerrillo
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Ignacio de Los Santos
- Infectious Diseases Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Jesús Sanz
- Infectious Diseases Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Lucio García-Fraile
- Infectious Diseases Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - Francisco Sánchez-Madrid
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| | - Arantzazu Alfranca
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa
| | - María Ángeles Muñoz-Fernández
- Immunology Section, Instituto Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón. Madrid, Spain
| | | | - Maria J Buzón
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona
| | - Alejandro Balazs
- Ragon Institute of MGH, MIT and Harvard.,Human Immune System Mouse Program from Massachusetts General Hospital, Boston
| | - Vladimir Vrbanac
- Ragon Institute of MGH, MIT and Harvard.,Human Immune System Mouse Program from Massachusetts General Hospital, Boston
| | - Enrique Martín-Gayo
- Immunology Unit from Hospital Universitario de la Princesa and Instituto de Investigación Sanitaria Princesa.,Universidad Autónoma of Madrid, Medicine Department Spain
| |
Collapse
|
9
|
Petkov S, Chiodi F. Distinct transcriptomic profiles of naïve CD4+ T cells distinguish HIV-1 infected patients initiating antiretroviral therapy at acute or chronic phase of infection. Genomics 2021; 113:3487-3500. [PMID: 34425224 DOI: 10.1016/j.ygeno.2021.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
We analyzed the whole transcriptome characteristics of blood CD4+ T naïve (TN) cells isolated from HIV-1 infected patients starting ART at acute (early ART = EA; n = 13) or chronic (late ART = LA; n = 11) phase of infection and controls (C; n = 15). RNA sequencing revealed 389 differentially expressed genes (DEGs) in EA and 810 in LA group in relation to controls. Comparison of the two groups of patients showed 183 DEGs. We focused on DEGs involved in apoptosis, inflammation and immune response. Clustering showed a poor separation of EA from C suggesting that these two groups present a similar transcriptomic profile of CD4+ TN cells. The comparison of EA and LA patients resulted in a high cluster purity revealing that different biological dysfunctions characterize EA and LA patients. The upregulated expression of several inflammatory chemokine genes distinguished the patient groups from C; CCL2 and CCL7, however, were downregulated in EA compared to LA patients. BCL2, an anti-apoptotic factor pivotal for naïve T cell homeostasis, distinguished both EA and LA from C. The expression of several DEGs involved in different inflammatory processes (TLR4, PTGS2, RAG1, IFNA16) was lower in EA compared LA. We conclude that although the transcriptome of CD4+ TN cells isolated from patients initiating ART at acute infection reveals a more quiescent phenotype, the survival profile of these cells still appears to be affected. Our results show that the detrimental process of inflammation is under more efficient control in EA patients.
Collapse
Affiliation(s)
- Stefan Petkov
- Department of Microbiology, Tumor and Cell Biology at Biomedicum, Karolinska Institutet, Solna, Sweden.
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology at Biomedicum, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
10
|
Armani-Tourret M, Zhou Z, Gasser R, Staropoli I, Cantaloube-Ferrieu V, Benureau Y, Garcia-Perez J, Pérez-Olmeda M, Lorin V, Puissant-Lubrano B, Assoumou L, Delaugerre C, Lelièvre JD, Lévy Y, Mouquet H, Martin-Blondel G, Alcami J, Arenzana-Seisdedos F, Izopet J, Colin P, Lagane B. Mechanisms of HIV-1 evasion to the antiviral activity of chemokine CXCL12 indicate potential links with pathogenesis. PLoS Pathog 2021; 17:e1009526. [PMID: 33872329 PMCID: PMC8084328 DOI: 10.1371/journal.ppat.1009526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/29/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
HIV-1 infects CD4 T lymphocytes (CD4TL) through binding the chemokine receptors CCR5 or CXCR4. CXCR4-using viruses are considered more pathogenic, linked to accelerated depletion of CD4TL and progression to AIDS. However, counterexamples to this paradigm are common, suggesting heterogeneity in the virulence of CXCR4-using viruses. Here, we investigated the role of the CXCR4 chemokine CXCL12 as a driving force behind virus virulence. In vitro, CXCL12 prevents HIV-1 from binding CXCR4 and entering CD4TL, but its role in HIV-1 transmission and propagation remains speculative. Through analysis of thirty envelope glycoproteins (Envs) from patients at different stages of infection, mostly treatment-naïve, we first interrogated whether sensitivity of viruses to inhibition by CXCL12 varies over time in infection. Results show that Envs resistant (RES) to CXCL12 are frequent in patients experiencing low CD4TL levels, most often late in infection, only rarely at the time of primary infection. Sensitivity assays to soluble CD4 or broadly neutralizing antibodies further showed that RES Envs adopt a more closed conformation with distinct antigenicity, compared to CXCL12-sensitive (SENS) Envs. At the level of the host cell, our results suggest that resistance is not due to improved fusion or binding to CD4, but owes to viruses using particular CXCR4 molecules weakly accessible to CXCL12. We finally asked whether the low CD4TL levels in patients are related to increased pathogenicity of RES viruses. Resistance actually provides viruses with an enhanced capacity to enter naive CD4TL when surrounded by CXCL12, which mirrors their situation in lymphoid organs, and to deplete bystander activated effector memory cells. Therefore, RES viruses seem more likely to deregulate CD4TL homeostasis. This work improves our understanding of the pathophysiology and the transmission of HIV-1 and suggests that RES viruses' receptors could represent new therapeutic targets to help prevent CD4TL depletion in HIV+ patients on cART.
Collapse
Affiliation(s)
| | - Zhicheng Zhou
- Viral Pathogenesis Unit, Department of Virology, INSERM U1108, Institut Pasteur, Paris, France
| | - Romain Gasser
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
| | - Isabelle Staropoli
- Viral Pathogenesis Unit, Department of Virology, INSERM U1108, Institut Pasteur, Paris, France
| | | | - Yann Benureau
- Viral Pathogenesis Unit, Department of Virology, INSERM U1108, Institut Pasteur, Paris, France
| | | | - Mayte Pérez-Olmeda
- AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III, Madrid, Spain
| | - Valérie Lorin
- Laboratory of Humoral Immunology, Department of Immunology, INSERM U1222, Institut Pasteur, Paris, France
| | | | - Lambert Assoumou
- INSERM, Sorbonne Université, Institut Pierre Louis d’Epidémiologie et de Santé Publique (IPLESP), Paris, France
| | | | | | - Yves Lévy
- Vaccine Research Institute, INSERM and APHP, Hôpital H. Mondor, Créteil, France
| | - Hugo Mouquet
- Laboratory of Humoral Immunology, Department of Immunology, INSERM U1222, Institut Pasteur, Paris, France
| | - Guillaume Martin-Blondel
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
- CHU de Toulouse, Service des Maladies Infectieuses et Tropicales, Toulouse, France
| | - Jose Alcami
- AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jacques Izopet
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
- CHU de Toulouse, Laboratoire de virologie, Toulouse, France
| | - Philippe Colin
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
| | - Bernard Lagane
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
- * E-mail:
| |
Collapse
|
11
|
Scholz EMB, Kashuba ADM. The Lymph Node Reservoir: Physiology, HIV Infection, and Antiretroviral Therapy. Clin Pharmacol Ther 2021; 109:918-927. [PMID: 33529355 DOI: 10.1002/cpt.2186] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/27/2021] [Indexed: 12/18/2022]
Abstract
Despite advances in treatment, finding a cure for HIV remains a top priority. Chronic HIV infection is associated with increased risk of comorbidities, such as diabetes and cardiovascular disease. Additionally, people living with HIV must remain adherent to daily antiretroviral therapy, because lapses in medication adherence can lead to viral rebound and disease progression. Viral recrudescence occurs from cellular reservoirs in lymphoid tissues. In particular, lymph nodes are central to the pathology of HIV due to their unique architecture and compartmentalization of immune cells. Understanding how antiretrovirals (ARVs) penetrate lymph nodes may explain why these tissues are maintained as HIV reservoirs, and how they contribute to viral rebound upon treatment interruption. In this report, we review (i) the physiology of the lymph nodes and their function as part of the immune and lymphatic systems, (ii) the pathogenesis and outcomes of HIV infection in lymph nodes, and (iii) ARV concentrations and distribution in lymph nodes, and the relationship between ARVs and HIV in this important reservoir.
Collapse
Affiliation(s)
- Erin M B Scholz
- Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina, USA
| | - Angela D M Kashuba
- Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina, USA.,School of Medicine, The University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
12
|
Li X, Zhao J, Kasinath V, Uehara M, Jiang L, Banouni N, McGrath MM, Ichimura T, Fiorina P, Lemos DR, Shin SR, Ware CF, Bromberg JS, Abdi R. Lymph node fibroblastic reticular cells deposit fibrosis-associated collagen following organ transplantation. J Clin Invest 2020; 130:4182-4194. [PMID: 32597832 PMCID: PMC7410068 DOI: 10.1172/jci136618] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/22/2020] [Indexed: 02/05/2023] Open
Abstract
Although the immune response within draining lymph nodes (DLNs) has been studied for decades, how their stromal compartment contributes to this process remains to be fully explored. Here, we show that donor mast cells were prominent activators of collagen I deposition by fibroblastic reticular cells (FRCs) in DLNs shortly following transplantation. Serial analysis of the DLN indicated that the LN stroma did not return to its baseline microarchitecture following organ rejection and that the DLN contained significant fibrosis following repetitive organ transplants. Using several FRC conditional-knockout mice, we show that induction of senescence in the FRCs of the DLN resulted in massive production of collagen I and a proinflammatory milieu within the DLN. Stimulation of herpes virus entry mediator (HVEM) on FRCs by its ligand LIGHT contributed chiefly to the induction of senescence in FRCs and overproduction of collagen I. Systemic administration of ex vivo-expanded FRCs to mice decreased DLN fibrosis and strengthened the effect of anti-CD40L in prolonging heart allograft survival. These data demonstrate that the transformation of FRCs into proinflammatory myofibroblasts is critically important for the maintenance of a proinflammatory milieu within a fibrotic DLN.
Collapse
Affiliation(s)
- Xiaofei Li
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei, China
| | - Jing Zhao
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Liwei Jiang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Naima Banouni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Martina M. McGrath
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei, China
| | | | - Paolo Fiorina
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dario R. Lemos
- Renal Division, Brigham and Women’s Hospital
- Harvard Stem Cell Institute, and
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl F. Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Jonathan S. Bromberg
- Department of Surgery and Microbiology and Immunobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Hillmann A, Crane M, Ruskin HJ. Assessing the impact of HIV treatment interruptions using stochastic cellular Automata. J Theor Biol 2020; 502:110376. [PMID: 32574568 DOI: 10.1016/j.jtbi.2020.110376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/23/2020] [Accepted: 06/12/2020] [Indexed: 11/30/2022]
Abstract
Chronic HIV infection causes a progressive decrease in the ability to maintain homeostasis resulting, after some time, in eventual break down of immune functions. Recent clinical research has shed light on a significant contribution of the lymphatic tissues, where HIV causes accumulation of collagen, (fibrosis). Specifically, where tissue is populated by certain types of functional stromal cells designated Fibroblastic Reticular Cells (FRCs), these have been found to play a crucial role in balancing out apoptosis and regeneration of naïve T-cells through 2-way cellular signaling. Tissue fibrosis not only impedes this signaling, effectively reducing T-cell levels through increased apoptosis of cells of both T- and FRC type but has been found to be irreversible by current HIV standard treatment (cART). While the therapy aims to block the viral lifecycle, cART-associated increase of T-cell levels in blood appears to conceal existing FRC impairment through fibrosis. This hidden impairment can lead to adverse consequences if treatment is interrupted, e.g. due to poor adherence (missing doses) or through periods recovering from drug toxicities. Formal clinical studies on treatment interruption have indicated possible adverse effects, but quantification of those effects in relation to interruption protocol and patient predisposition remains unclear. Accordingly, the impact of treatment interruption on lymphatic tissue structure and T-cell levels is explored here by means of computer simulation. A novel Stochastic Cellular Automata model is proposed, which utilizes all sources of clinical detail available to us (though sparse in part) for model parametrization. Sources are explicitly referenced and conflicting evidence from previous studies explored. The main focus is on (i) spatial aspects of collagen build up, together with (ii) collagen increase after repeated treatment interruptions to explore the dynamics of HIV-induced fibrosis and T-cell loss.
Collapse
Affiliation(s)
- Andreas Hillmann
- Advanced Research Computing Centre for Complex Systems Modelling, School of Computing, Dublin City University, Dublin, Ireland.
| | - Martin Crane
- Advanced Research Computing Centre for Complex Systems Modelling, School of Computing, Dublin City University, Dublin, Ireland
| | - Heather J Ruskin
- Advanced Research Computing Centre for Complex Systems Modelling, School of Computing, Dublin City University, Dublin, Ireland
| |
Collapse
|
14
|
McCarthy MK, Reynoso GV, Winkler ES, Mack M, Diamond MS, Hickman HD, Morrison TE. MyD88-dependent influx of monocytes and neutrophils impairs lymph node B cell responses to chikungunya virus infection via Irf5, Nos2 and Nox2. PLoS Pathog 2020; 16:e1008292. [PMID: 31999809 PMCID: PMC7012455 DOI: 10.1371/journal.ppat.1008292] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 02/11/2020] [Accepted: 12/22/2019] [Indexed: 12/21/2022] Open
Abstract
Humoral immune responses initiate in the lymph node draining the site of viral infection (dLN). Some viruses subvert LN B cell activation; however, our knowledge of viral hindrance of B cell responses of important human pathogens is lacking. Here, we define mechanisms whereby chikungunya virus (CHIKV), a mosquito-transmitted RNA virus that causes outbreaks of acute and chronic arthritis in humans, hinders dLN antiviral B cell responses. Infection of WT mice with pathogenic, but not acutely cleared CHIKV, induced MyD88-dependent recruitment of monocytes and neutrophils to the dLN. Blocking this influx improved lymphocyte accumulation, dLN organization, and CHIKV-specific B cell responses. Both inducible nitric oxide synthase (iNOS) and the phagocyte NADPH oxidase (Nox2) contributed to impaired dLN organization and function. Infiltrating monocytes expressed iNOS through a local IRF5- and IFNAR1-dependent pathway that was partially TLR7-dependent. Together, our data suggest that pathogenic CHIKV triggers the influx and activation of monocytes and neutrophils in the dLN that impairs virus-specific B cell responses. Elucidating mechanisms by which viruses subvert B cell immunity and establish persistent infection is essential for the development of new therapeutic strategies against chronic viral infections. The humoral immune response initiates in the lymph node draining the site of viral infection. However, how persistent viruses evade B cell responses is poorly understood. In this study, we find that infection with pathogenic, persistent chikungunya virus triggers rapid recruitment of neutrophils and monocytes to the draining lymph node, which impair structural organization, lymphocyte accumulation, and downstream virus-specific B cell responses that are important for control of infection. This work enhances our understanding of the pathogenesis of acute and chronic CHIKV disease and highlights how local innate immune responses in draining lymphoid tissue dictate the effectiveness of downstream adaptive immunity.
Collapse
Affiliation(s)
- Mary K. McCarthy
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Glennys V. Reynoso
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Microbiology and Immunology, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Emma S. Winkler
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Matthias Mack
- Regensburg University Medical Center, Regensburg, Germany
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Heather D. Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Microbiology and Immunology, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
15
|
Saxena V, Li L, Paluskievicz C, Kasinath V, Bean A, Abdi R, Jewell CM, Bromberg JS. Role of lymph node stroma and microenvironment in T cell tolerance. Immunol Rev 2019; 292:9-23. [PMID: 31538349 PMCID: PMC6935411 DOI: 10.1111/imr.12799] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022]
Abstract
Lymph nodes (LNs) are at the cross roads of immunity and tolerance. These tissues are compartmentalized into specialized niche areas by lymph node stromal cells (LN SCs). LN SCs shape the LN microenvironment and guide immunological cells into different zones through establishment of a CCL19 and CCL21 gradient. Following local immunological cues, LN SCs modulate activity to support immune cell priming, activation, and fate. This review will present our current understanding of LN SC subsets roles in regulating T cell tolerance. Three major types of LN SC subsets, namely fibroblastic reticular cells, lymphatic endothelial cells, and blood endothelial cells, are discussed. These subsets serve as scaffolds to support and regulate T cell homeostasis. They contribute to tolerance by presenting peripheral tissue antigens to both CD4 and CD8 T cells. The role of LN SCs in regulating T cell migration and tolerance induction is discussed. Looking forward, recent advances in bioengineered materials and approaches to leverage LN SCs to induce T cell tolerance are highlighted, as are current clinical practices that allow for manipulation of the LN microenvironment to induce tolerance. Increased understanding of LN architecture, how different LN SCs integrate immunological cues and shape immune responses, and approaches to induce T cell tolerance will help further combat autoimmune diseases and graft rejection.
Collapse
Affiliation(s)
- Vikas Saxena
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lushen Li
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Christina Paluskievicz
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Vivek Kasinath
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Asher Bean
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Reza Abdi
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, Robert E. Fischell Institute for Biomedical Devices University of Maryland, College Park, MD 20742, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Jonathan S. Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Furler RL, Newcombe KL, Del Rio Estrada PM, Reyes-Terán G, Uittenbogaart CH, Nixon DF. Histoarchitectural Deterioration of Lymphoid Tissues in HIV-1 Infection and in Aging. AIDS Res Hum Retroviruses 2019; 35:1148-1159. [PMID: 31474115 DOI: 10.1089/aid.2019.0156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Impaired immunity is a common symptom of aging and advanced Human Immunodeficiency Virus type 1 (HIV-1) disease. In both diseases, a decline in lymphocytic function and cellularity leads to ineffective adaptive immune responses to opportunistic infections and vaccinations. Furthermore, despite sustained myeloid cellularity there is a background of chronic immune activation and a decrease in innate immune function in aging. In HIV-1 disease, myeloid cellularity is often more skewed than in normal aging, but similar chronic activation and innate immune dysfunction typically arise. Similarities between aging and HIV-1 infection have led to several investigations into HIV-1-mediated aging of the immune system. In this article, we review various studies that report alterations of leukocyte number and function during aging, and compare those alterations with those observed during progressive HIV-1 disease. We pay particular attention to changes within lymphoid tissue microenvironments and how histoarchitectural changes seen in these two diseases affect immunity. As we review various immune compartments including peripheral blood as well as primary and secondary lymphoid organs, common themes arise that help explain the decline of immunity in the elderly and in HIV-1-infected individuals with advanced disease. In both conditions, lymphoid tissues often show signs of histoarchitectural deterioration through fat accumulation and/or fibrosis. These structural changes can be attributed to a loss of communication between leukocytes and the surrounding stromal cells that produce the extracellular matrix components and growth factors necessary for cell migration, cell proliferation, and lymphoid tissue function. Despite the common general impairment of immunity in aging and HIV-1 progression, deterioration of immunity is caused by distinct mechanisms at the cellular and tissue levels in these two diseases.
Collapse
Affiliation(s)
- Robert L. Furler
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Kevin L. Newcombe
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Perla M. Del Rio Estrada
- Departmento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” CDMX, Mexico DF, Mexico
| | - Gustavo Reyes-Terán
- Departmento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” CDMX, Mexico DF, Mexico
| | - Christel H. Uittenbogaart
- Department of Microbiology, Immunology and Molecular Genetics, Medicine-Pediatrics, UCLA AIDS Institute and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - Douglas F. Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
17
|
Lofano G, Gorman MJ, Yousif AS, Yu WH, Fox JM, Dugast AS, Ackerman ME, Suscovich TJ, Weiner J, Barouch D, Streeck H, Little S, Smith D, Richman D, Lauffenburger D, Walker BD, Diamond MS, Alter G. Antigen-specific antibody Fc glycosylation enhances humoral immunity via the recruitment of complement. Sci Immunol 2019; 3:3/26/eaat7796. [PMID: 30120121 PMCID: PMC6298214 DOI: 10.1126/sciimmunol.aat7796] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/29/2018] [Indexed: 12/14/2022]
Abstract
HIV-specific broadly neutralizing antibodies (bNAbs) confer protection after passive immunization, but the immunological mechanisms that drive their development are poorly understood. Structural features of bNAbs indicate that they originate from extensive germinal center (GC) selection, which relies on persistent GC activity. However, why a fraction of infected individuals are able to successfully drive more effective affinity maturation is unclear. Delivery of antigens in the form of antibody-immune complexes (ICs), which bind to complement receptors (CRs) or Fc receptors (FcRs) on follicular dendritic cells, represents an effective mechanism for antigen delivery to the GC. We sought to define whether IC-FcR or CR interactions differ among individuals who develop bNAb responses to HIV. Enhanced Fc effector functions and FcR/CR interactions, via altered Fc glycosylation profiles, were observed among individuals with neutralizing antibody responses to HIV compared with those without neutralizing antibody activity. Moreover, both polyclonal neutralizer ICs and monoclonal IC mimics of neutralizer antibodies induced higher antibody titers, higher-avidity antibodies, and expanded GC B cell reactions after immunization of mice via accelerated antigen deposition within B cell follicles in a complement-dependent manner. Thus, these data point to a direct role for altered Fc profile/complement interactions in shaping the maturation of the humoral immune response, providing insights into how GC activity may be enhanced to drive affinity maturation in next-generation vaccine approaches.
Collapse
Affiliation(s)
- Giuseppe Lofano
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Matthew J Gorman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ashraf S Yousif
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.,Department of Immunology and Biotechnology, Tropical Medicine Research Institute, Khartoum, Sudan
| | - Wen-Han Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Julie M Fox
- Departments of Medicine, Molecular Microbiology, and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | - Todd J Suscovich
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Joshua Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Dan Barouch
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.,Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Hendrik Streeck
- Institut für HIV Forschung, Universität Duisburg-Essen, Essen, Germany
| | - Susan Little
- University of California, San Diego, San Diego, CA 92093, USA
| | - Davey Smith
- University of California, San Diego, San Diego, CA 92093, USA.,VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Douglas Richman
- University of California, San Diego, San Diego, CA 92093, USA.,VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
18
|
Poultsidi A, Dimopoulos Y, He TF, Chavakis T, Saloustros E, Lee PP, Petrovas C. Lymph Node Cellular Dynamics in Cancer and HIV: What Can We Learn for the Follicular CD4 (Tfh) Cells? Front Immunol 2018; 9:2233. [PMID: 30319664 PMCID: PMC6170630 DOI: 10.3389/fimmu.2018.02233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/07/2018] [Indexed: 12/17/2022] Open
Abstract
Lymph nodes (LNs) are central in the generation of adaptive immune responses. Follicular helper CD4 T (Tfh) cells, a highly differentiated CD4 population, provide critical help for the development of antigen-specific B cell responses within the germinal center. Throughout the past decade, numerous studies have revealed the important role of Tfh cells in Human Immunodeficiency Virus (HIV) pathogenesis as well as in the development of neutralizing antibodies post-infection and post-vaccination. It has also been established that tumors influence various immune cell subsets not only in their proximity, but also in draining lymph nodes. The role of local or tumor associated lymph node Tfh cells in disease progression is emerging. Comparative studies of Tfh cells in chronic infections and cancer could therefore provide novel information with regards to their differentiation plasticity and to the mechanisms regulating their development.
Collapse
Affiliation(s)
- Antigoni Poultsidi
- Department of Surgery, Medical School, University of Thessaly, Larissa, Greece
| | - Yiannis Dimopoulos
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| | - Ting-Fang He
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Emmanouil Saloustros
- Department of Internal Medicine, Medical School, University of Thessaly, Larissa, Greece
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| |
Collapse
|
19
|
Samal J, Kelly S, Na-Shatal A, Elhakiem A, Das A, Ding M, Sanyal A, Gupta P, Melody K, Roland B, Ahmed W, Zakir A, Bility M. Human immunodeficiency virus infection induces lymphoid fibrosis in the BM-liver-thymus-spleen humanized mouse model. JCI Insight 2018; 3:120430. [PMID: 30232273 DOI: 10.1172/jci.insight.120430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022] Open
Abstract
A major pathogenic feature associated with HIV infection is lymphoid fibrosis, which persists during antiretroviral therapy (ART). Lymphoid tissues play critical roles in the generation of antigen-specific immune response, and fibrosis disrupts the stromal network of lymphoid tissues, resulting in impaired immune cell trafficking and function, as well as immunodeficiency. Developing an animal model for investigating the impact of HIV infection-induced lymphoid tissue fibrosis on immunodeficiency and immune cell impairment is critical for therapeutics development and clinical translation. Said model will enable in vivo mechanistic studies, thus complementing the well-established surrogate model of SIV infection-induced lymphoid tissue fibrosis in macaques. We developed a potentially novel human immune system-humanized mouse model by coengrafting autologous fetal thymus, spleen, and liver organoids under the kidney capsule, along with i.v. injection of autologous fetal liver-derived hematopoietic stem cells, thus termed the BM-liver-thymus-spleen (BLTS) humanized mouse model. BLTS humanized mouse model supports development of human immune cells and human lymphoid organoids (human thymus and spleen organoids). HIV infection in BLTS humanized mice results in progressive fibrosis in human lymphoid tissues, which was associated with immunodeficiency in the lymphoid tissues, and lymphoid tissue fibrosis persists during ART, thus recapitulating clinical outcomes.
Collapse
|
20
|
Murakami T, Kim J, Li Y, Green GE, Shikanov A, Ono A. Secondary lymphoid organ fibroblastic reticular cells mediate trans-infection of HIV-1 via CD44-hyaluronan interactions. Nat Commun 2018; 9:2436. [PMID: 29934525 PMCID: PMC6015004 DOI: 10.1038/s41467-018-04846-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 05/22/2018] [Indexed: 12/21/2022] Open
Abstract
Fibroblastic reticular cells (FRCs) are stromal cells in secondary lymphoid organs, the major sites for HIV-1 infection of CD4+ T cells. Although FRCs regulate T cell survival, proliferation, and migration, whether they play any role in HIV-1 spread has not been studied. Here, we show that FRCs enhance HIV-1 spread via trans-infection in which FRCs capture HIV-1 and facilitate infection of T cells that come into contact with FRCs. FRCs mediate trans-infection in both two- and three-dimensional culture systems and in a manner dependent on the virus producer cells. This producer cell dependence, which was also observed for virus spread in secondary lymphoid tissues ex vivo, is accounted for by CD44 incorporated into virus particles and hyaluronan bound to such CD44 molecules. This virus-associated hyaluronan interacts with CD44 expressed on FRCs, thereby promoting virus capture by FRCs. Overall, our results reveal a novel role for FRCs in promoting HIV-1 spread. Fibroblastic reticular cells (FRCs) are important regulators of T cell survival, proliferation, and migration in secondary lymphoid organs, but their role in HIV infection isn’t studied. Here, Murakami et al. show that FRCs enhance HIV spread via CD44- and hyaluronan-mediated trans-infection.
Collapse
Affiliation(s)
- Tomoyuki Murakami
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Jiwon Kim
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yi Li
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Glenn Edward Green
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Ariella Shikanov
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
21
|
Aid M, Dupuy FP, Moysi E, Moir S, Haddad EK, Estes JD, Sekaly RP, Petrovas C, Ribeiro SP. Follicular CD4 T Helper Cells As a Major HIV Reservoir Compartment: A Molecular Perspective. Front Immunol 2018; 9:895. [PMID: 29967602 PMCID: PMC6015877 DOI: 10.3389/fimmu.2018.00895] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/10/2018] [Indexed: 01/05/2023] Open
Abstract
Effective antiretroviral therapy (ART) has prevented the progression to AIDS and reduced HIV-related morbidities and mortality for the majority of infected individuals. However, a lifelong administration of ART is necessary, placing an inordinate burden on individuals and public health systems. Therefore, discovering therapeutic regimens able to eradicate or functionally cure HIV infection is of great importance. ART interruption leads to viral rebound highlighting the establishment and maintenance of a latent viral reservoir compartment even under long-term treatment. Follicular helper CD4 T cells (TFH) have been reported as a major cell compartment contributing to viral persistence, consequent to their susceptibility to infection and ability to release replication-competent new virions. Here, we discuss the molecular profiles and potential mechanisms that support the role of TFH cells as one of the major HIV reservoirs.
Collapse
Affiliation(s)
- Malika Aid
- Beth Israel Deaconess Medical Center, Center for Virology and Vaccine Research, Harvard Medical School, Boston, MA, United States
| | - Frank P Dupuy
- Centre hospitalier de l'Université de Montréal, Montreal, QC, United States
| | - Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIH, Bethesda, MD, United States
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, United States
| | - Elias K Haddad
- Division of Infectious Diseases & HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jacob D Estes
- Oregon National Primate Research Center, Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Rafick Pierre Sekaly
- Pathology Department, Case Western Reserve University, Cleveland, OH, United States
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIH, Bethesda, MD, United States
| | | |
Collapse
|
22
|
Ferrando-Martinez S, Moysi E, Pegu A, Andrews S, Nganou Makamdop K, Ambrozak D, McDermott AB, Palesch D, Paiardini M, Pavlakis GN, Brenchley JM, Douek D, Mascola JR, Petrovas C, Koup RA. Accumulation of follicular CD8+ T cells in pathogenic SIV infection. J Clin Invest 2018; 128:2089-2103. [PMID: 29664020 DOI: 10.1172/jci96207] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 03/06/2018] [Indexed: 01/06/2023] Open
Abstract
LN follicles constitute major reservoir sites for HIV/SIV persistence. Cure strategies could benefit from the characterization of CD8+ T cells able to access and eliminate HIV-infected cells from these areas. In this study, we provide a comprehensive analysis of the phenotype, frequency, localization, and functionality of follicular CD8+ T cells (fCD8+) in SIV-infected nonhuman primates. Although disorganization of follicles was a major factor, significant accumulation of fCD8+ cells during chronic SIV infection was also observed in intact follicles, but only in pathogenic SIV infection. In line with this, tissue inflammatory mediators were strongly associated with the accumulation of fCD8+ cells, pointing to tissue inflammation as a major factor in this process. These fCD8+ cells have cytolytic potential and can be redirected to target and kill HIV-infected cells using bispecific antibodies. Altogether, our data support the use of SIV infection to better understand the dynamics of fCD8+ cells and to develop bispecific antibodies as a strategy for virus eradication.
Collapse
Affiliation(s)
| | | | | | | | - Krystelle Nganou Makamdop
- Human Immunology Section, Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | | | | | - David Palesch
- Department of Pathology, Emory University School of Medicine and Yerkes National Primate Research Center, Atlanta, Georgia, USA
| | - Mirko Paiardini
- Department of Pathology, Emory University School of Medicine and Yerkes National Primate Research Center, Atlanta, Georgia, USA
| | - George N Pavlakis
- Human Retrovirus Section, Center for Cancer Research, National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Daniel Douek
- Human Immunology Section, Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
23
|
Jackson L, Hunter J, Cele S, Ferreira IM, Young AC, Karim F, Madansein R, Dullabh KJ, Chen CY, Buckels NJ, Ganga Y, Khan K, Boulle M, Lustig G, Neher RA, Sigal A. Incomplete inhibition of HIV infection results in more HIV infected lymph node cells by reducing cell death. eLife 2018; 7:30134. [PMID: 29555018 PMCID: PMC5896883 DOI: 10.7554/elife.30134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 03/08/2018] [Indexed: 12/22/2022] Open
Abstract
HIV has been reported to be cytotoxic in vitro and in lymph node infection models. Using a computational approach, we found that partial inhibition of transmissions of multiple virions per cell could lead to increased numbers of live infected cells. If the number of viral DNA copies remains above one after inhibition, then eliminating the surplus viral copies reduces cell death. Using a cell line, we observed increased numbers of live infected cells when infection was partially inhibited with the antiretroviral efavirenz or neutralizing antibody. We then used efavirenz at concentrations reported in lymph nodes to inhibit lymph node infection by partially resistant HIV mutants. We observed more live infected lymph node cells, but with fewer HIV DNA copies per cell, relative to no drug. Hence, counterintuitively, limited attenuation of HIV transmission per cell may increase live infected cell numbers in environments where the force of infection is high. The HIVvirus infects cells of the immune system. Once inside, it hijacks the cellular molecular machineries to make more copies of itself, which are then transmitted to new host cells. HIV eventually kills most cells it infects, either in the steps leading to the infection of the cell, or after the cell is already producing virus. HIV can spread between cells in two ways, known as cell-free or cell-to-cell. In the first, individual viruses are released from infected cells and move randomly through the body in the hope of finding new cells to infect. In the second, infected cells interact directly with uninfected cells. The second method is often much more successful at infecting new cells since they are exposed to multiple virus particles. HIV infections can be controlled by using combinations of antiretroviral drugs, such as efavirenz, to prevent the virus from making more of itself. With a high enough dose, the drugs can in theory completely stop HIV infections, unless the virus becomes resistant to treatment. However, some patients continue to use these drugs even after the virus they are infected with develops resistance. It is not clear what effect taking ineffective, or partially effective, drugs has on how HIV progresses. Using efavirenz, Jackson, Hunter et al. partially limited the spread of HIV between human cells grown in the laboratory. The experiments mirrored the situation where a partially resistant HIV strain spreads through the body. The results show that the success of cell-free infection is reduced as drug dose increases. Yet paradoxically, in cell-to-cell infection, the presence of drug caused more cells to become infected. This can be explained by the fact that, in cell-to-cell spread, each cell is exposed to multiple copies of the virus. The drug dose reduced the number of viral copies per cell without stopping the virus from infecting completely. The reduced number of viral copies per cell made it more likely that infected cells would survive the infection long enough to produce virus particles themselves. Viruses that can kill cells, such as HIV, must balance the need to make more of themselves against the speed that they kill their host cell to maximize the number of infected cells. If transmission between cells is too effective and too many virus particles are delivered to the new cell, the virus may not manage to infect new hosts before killing the old ones. These findings highlight this delicate balance. They also indicate a potential issue in using drugs to treat partially resistant virus strains. Without care, these treatments could increase the number of infected cells in the body, potentially worsening the effects of living with HIV.
Collapse
Affiliation(s)
- Laurelle Jackson
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Jessica Hunter
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sandile Cele
- Africa Health Research Institute, Durban, South Africa
| | - Isabella Markham Ferreira
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andrew C Young
- Africa Health Research Institute, Durban, South Africa.,Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, United States
| | - Farina Karim
- Africa Health Research Institute, Durban, South Africa
| | - Rajhmun Madansein
- Department of Cardiothoracic Surgery, University of KwaZulu-Natal, Durban, South Africa.,Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Kaylesh J Dullabh
- Department of Cardiothoracic Surgery, University of KwaZulu-Natal, Durban, South Africa
| | - Chih-Yuan Chen
- Department of Cardiothoracic Surgery, University of KwaZulu-Natal, Durban, South Africa
| | - Noel J Buckels
- Department of Cardiothoracic Surgery, University of KwaZulu-Natal, Durban, South Africa
| | - Yashica Ganga
- Africa Health Research Institute, Durban, South Africa
| | - Khadija Khan
- Africa Health Research Institute, Durban, South Africa
| | - Mikael Boulle
- Africa Health Research Institute, Durban, South Africa
| | - Gila Lustig
- Africa Health Research Institute, Durban, South Africa
| | - Richard A Neher
- Biozentrum, University of Basel, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.,Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
24
|
Abstract
Lymph nodes play a central role in the development of adaptive immunity against pathogens and particularly the generation of antigen-specific B cell responses in specialized areas called germinal centers (GCs). Lymph node (LN) pathology was recognized as an important consequence of human immunodeficiency virus (HIV) infection since the beginning of the HIV epidemic. Investigation into the structural and functional alterations induced by HIV and Simian immunodeficiency virus (SIV) has further cemented the central role that lymphoid tissue plays in HIV/SIV pathogenesis. The coexistence of constant local inflammation, altered tissue architecture, and relative exclusion of virus-specific CD8 T cells from the GCs creates a unique environment for the virus evolution and establishment of viral reservoir in specific GC cells, namely T follicular helper CD4 T cells (Tfh). A better understanding of the biology of immune cells in HIV-infected lymph nodes is a prerequisite to attaining the ultimate goal of complete viral eradication.
Collapse
Affiliation(s)
- Yiannis Dimopoulos
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, 40 Convent Drive, MSC 3022, Building 40, Room 3612B, Bethesda, MD, 20892, USA
| | - Eirini Moysi
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, 40 Convent Drive, MSC 3022, Building 40, Room 3612B, Bethesda, MD, 20892, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, 40 Convent Drive, MSC 3022, Building 40, Room 3612B, Bethesda, MD, 20892, USA.
| |
Collapse
|
25
|
Li Y, Sun W. Effects of Th17/Treg cell imbalance on HIV replication in patients with AIDS complicated with tuberculosis. Exp Ther Med 2018; 15:2879-2883. [PMID: 29456692 PMCID: PMC5795545 DOI: 10.3892/etm.2018.5768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 09/04/2017] [Indexed: 12/13/2022] Open
Abstract
The purpose of this study was to determine the effect of Th17/Treg cell imbalance on HIV replication in patients with AIDS complicated with tuberculosis (TB). We selected 32 patients with AIDS combined with TB infection in our hospital and 30 healthy individual as controls. The Th17/Treg ratio in peripheral blood lymphocytes was detected by flow cytometry. Compared with healthy subjects, Th17 cells first declined in HIV patients with TB, but gradually increased over the course of the disease. Treg showed an increasing trend in HIV patients with TB. The Th17/Treg ratio was significantly altered as the condition gradually deteriorated. ELISA showed that interleukin (IL)-17, IL-6 and IL-10 in patients with HIV complicated with TB were significantly lower than in healthy subjects. The imbalance of Th17/Treg cells can promote HIV virus replication in AIDS patients with TB infection, which can aggravate the condition.
Collapse
Affiliation(s)
- Yanshuang Li
- Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Weijia Sun
- Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
26
|
Kaye PM. Stromal Cell Responses in Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:23-36. [DOI: 10.1007/978-3-319-78127-3_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Vella LA, Herati RS, Wherry EJ. CD4 + T Cell Differentiation in Chronic Viral Infections: The Tfh Perspective. Trends Mol Med 2017; 23:1072-1087. [PMID: 29137933 PMCID: PMC5886740 DOI: 10.1016/j.molmed.2017.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/11/2017] [Accepted: 10/16/2017] [Indexed: 12/18/2022]
Abstract
CD4+ T cells play a critical role in the response to chronic viral infections during the acute phase and in the partial containment of infections once chronic infection is established. As infection persists, the virus-specific CD4+ T cell response begins to shift in phenotype. The predominant change described in both mouse and human studies of chronic viral infection is a decrease in detectable T helper type (Th)1 responses. Some Th1 loss is due to decreased proliferative potential and decreased cytokine production in the setting of chronic antigen exposure. However, recent data suggest that Th1 dysfunction is accompanied by a shift in the differentiation pathway of virus-specific CD4+ T cells, with enrichment for cells with a T follicular helper cell (Tfh) phenotype. A Tfh-like program during chronic infection has now been identified in virus-specific CD8+ T cells as well. In this review, we discuss what is known about CD4+ T cell differentiation in chronic viral infections, with a focus on the emergence of the Tfh program and the implications of this shift with respect to Tfh function and the host-pathogen interaction.
Collapse
Affiliation(s)
- Laura A Vella
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Ramin S Herati
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
28
|
De Boer RJ, Perelson AS. How Germinal Centers Evolve Broadly Neutralizing Antibodies: the Breadth of the Follicular Helper T Cell Response. J Virol 2017; 91:e00983-17. [PMID: 28878083 PMCID: PMC5660473 DOI: 10.1128/jvi.00983-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/11/2017] [Indexed: 12/20/2022] Open
Abstract
Many HIV-1-infected patients evolve broadly neutralizing antibodies (bnAbs). This evolutionary process typically takes several years and is poorly understood as selection taking place in germinal centers occurs on the basis of antibody affinity. B cells with the highest-affinity receptors tend to acquire the most antigen from the follicular dendritic cell (FDC) network and present the highest density of cognate peptides to follicular helper T (Tfh) cells, which provide survival signals to the B cell. bnAbs are therefore expected to evolve only when the B cell lineage evolving breadth is consistently capturing and presenting more peptides to Tfh cells than other lineages of more specific B cells. Here we develop mathematical models of Tfh cells in germinal centers to explicitly define the mechanisms of selection in this complex evolutionary process. Our results suggest that broadly reactive B cells presenting a high density of peptides bound to major histocompatibility complex class II molecules (pMHC) are readily outcompeted by B cells responding to lineages of HIV-1 that transiently dominate the within host viral population. Conversely, if broadly reactive B cells acquire a large variety of several HIV-1 proteins from the FDC network and present a high diversity of several pMHC, they can be rescued by a large fraction of the Tfh cell repertoire in the germinal center. Under such circumstances the evolution of bnAbs is much more consistent. Increasing either the magnitude of the Tfh cell response or the breadth of the Tfh cell repertoire markedly facilitates the evolution of bnAbs. Because both the magnitude and breadth can be increased by vaccination with several HIV-1 proteins, this calls for experimental testing.IMPORTANCE Many HIV-infected patients slowly evolve antibodies that can neutralize a large variety of viruses. Such broadly neutralizing antibodies (bnAbs) could in the future become therapeutic agents. bnAbs appear very late, and patients are typically not protected by them. At the moment, we fail to understand why this takes so long and how the immune system selects for broadly neutralizing capacity. Typically, antibodies are selected based on affinity and not on breadth. We developed mathematical models to study two different mechanisms by which the immune system can select for broadly neutralizing capacity. One of these is based upon the repertoire of different follicular helper T (Tfh) cells in germinal centers. We suggest that broadly reactive B cells may interact with a larger fraction of this repertoire and demonstrate that this would select for bnAbs. Intriguingly, this suggests that broadening the Tfh cell repertoire by vaccination may speed up the evolution of bnAbs.
Collapse
Affiliation(s)
- Rob J De Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, The Netherlands
- Santa Fe Institute, Santa Fe, New Mexico, USA
| | - Alan S Perelson
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
- Santa Fe Institute, Santa Fe, New Mexico, USA
| |
Collapse
|
29
|
Gao J, Zhao L, Liu L, Yang Y, Guo B, Zhu B. Disrupted fibroblastic reticular cells and interleukin-7 expression in tumor draining lymph nodes. Oncol Lett 2017; 14:2954-2960. [PMID: 28928833 PMCID: PMC5588138 DOI: 10.3892/ol.2017.6537] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/28/2017] [Indexed: 11/06/2022] Open
Abstract
The immune system of patients with cancer is usually in an inhibitory state. Lymph node (LN) draining of pathological sites provides a suitable microenvironment where adaptive immune responses mainly occur. However, the microenvironment in the tumor draining lymph nodes (TDLNs) of patients with cancer appears to be in favor of tolerance. The effects of tumor cells on TDLNs have not been elaborated clearly. The present results have indicated that tumor cells may directly affect TDLNs by decreasing the fibroblastic reticular cell population that led to less interleukin-7 secretion. As a result, the number of T cells in TDLNs declined with reduced survival signals. A decreased number of T cells in TDLNs means weakened ability of immune surveillance. Clinically, these results were also confirmed in LN biopsies from patients with colon cancer at different clinical stages. Results of the present study showed that tumor cells may directly inhibit the immunological function of TDLNs.
Collapse
Affiliation(s)
- Jianbao Gao
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Lintao Zhao
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China.,Department of Oncology, The People's Liberation Army No. 324 Hospital, Chongqing 404000, P.R. China
| | - Lina Liu
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yang Yang
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Bo Guo
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China.,Department of Pathogenic Biology, Third Military Medical University, Chongqing 400038, P.R. China
| | - Bo Zhu
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
30
|
Sun Y, Fu Y, Zhang Z, Tang T, Liu J, Ding H, Han X, Xu J, Chu Z, Shang H, Jiang Y. The investigation of CD4+T-cell functions in primary HIV infection with antiretroviral therapy. Medicine (Baltimore) 2017; 96:e7430. [PMID: 28700479 PMCID: PMC5515751 DOI: 10.1097/md.0000000000007430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection leads to reduced CD4T-cell counts and immune dysfunction. Initiation of antiretroviral therapy (ART) in HIV primary infection has been recommended to achieve an optimal clinical outcome, but a comprehensive study on restoration of CD4T-cell function in primary HIV-infected individuals with ART still needs to be eluciated. We investigated longitudinal changes in the CD4T-cell counts, phenotypes, and functions in HIV-infected individuals with early ART (initiated within 6 months after HIV infection) or later ART (initiated more than 12 months after HIV infection). Patients from early ART and later ART groups had received ART for at least 1 year. Individuals with early ART had more CD4T cells, a faster rate of CD4T-cell recovery than those receiving later ART; the levels of CD4T-cell activation and senescence were lower in early ART compared to those with later ART (P = .031; P = .016), but the activation was higher than normal controls (NC) (P = .001); thymic emigrant function was more upregulated in early ART than in later ART (P = .015), but still lower than NC (P = .027); proliferative capacity and interferon-γ secretion of CD4T cells were significantly decreased in primary infection (P < .001; P = .029), and early ART restored these CD4T-cell functions, there is no difference with NC, later ART could partially restore the functions of CD4T cells, but it remained lower than that of NC (P = .005; P = .019). Early ART could better improve CD4T-cell function.
Collapse
|
31
|
Production of IgG antibodies to pneumococcal polysaccharides is associated with expansion of ICOS+ circulating memory T follicular-helper cells which is impaired by HIV infection. PLoS One 2017; 12:e0176641. [PMID: 28463977 PMCID: PMC5413043 DOI: 10.1371/journal.pone.0176641] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/13/2017] [Indexed: 11/26/2022] Open
Abstract
Dysfunction of T follicular-helper (TFH) cells is a possible cause of impaired germinal centre (GC) and IgG antibody responses in individuals with human immunodeficiency virus-1 (HIV-1) infection and might contribute to decreased magnitude and isotype diversification of IgG antibodies to pneumococcal polysaccharides (PcPs). We examined the production of IgG1 and IgG2 antibodies to PcPs 4, 6B, 9V and 14 by enumerating antibody secreting cells (ASCs) at day (D) 7 and determining fold-increase in serum antibody levels at D28 after vaccination with unconjugated PcPs in HIV seronegative subjects (n = 20) and in HIV patients who were receiving antiretroviral therapy (ART) (n = 28) or who were ART-naive (n = 11) and determined their association with ICOS+ and ICOS- circulating memory TFH (cmTFH) cells (CD4+CD45RA-CD27+CXCR5+PD-1+) and short lived plasmablasts (SPBs) at D7, and with PcP-specific and total IgM+ and IgG+ memory B cells at D0. In HIV seronegative subjects, production of IgG1+ and IgG2+ ASCs was consistently associated with the frequency of ICOS+ cmTFH cells but not ICOS- cmTFH cells or memory B cells. In contrast, post-vaccination ASCs in HIV patients, regardless of ART status, were lower than in HIV seronegative subjects and not associated with ICOS+ cmTFH cells, the expansion of which was absent (ART-naive patients) or much lower than in HIV seronegative subjects (ART-treated patients). Production of SPBs was also lower in ART-naive patients. Fold-increase in IgG2 antibodies at D28 also correlated with ICOS+ cmTFH cells at D7 in HIV seronegative subjects but not in HIV patients. These novel findings provide evidence that ICOS+ cmTFH cells contribute to the regulation of PcP-specific IgG antibody responses, including isotype diversification, and that TFH cell dysfunction may be a cause of impaired PcP-specific IgG antibody responses and increased susceptibility to pneumococcal disease in HIV patients.
Collapse
|
32
|
Ansari AR, Liu H. Acute Thymic Involution and Mechanisms for Recovery. Arch Immunol Ther Exp (Warsz) 2017; 65:401-420. [PMID: 28331940 DOI: 10.1007/s00005-017-0462-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 03/12/2017] [Indexed: 12/14/2022]
Abstract
Acute thymic involution (ATI) is usually regarded as a virulence trait. It is caused by several infectious agents (bacteria, viruses, parasites, fungi) and other factors, including stress, pregnancy, malnutrition and chemotherapy. However, the complex mechanisms that operate during ATI differ substantially from each other depending on the causative agent. For instance, a transient reduction in the size and weight of the thymus and depletion of populations of T cell subsets are hallmarks of ATI in many cases, whereas severe disruption of the anatomical structure of the organ is also associated with some factors, including fungal, parasitic and viral infections. However, growing evidence shows that ATI may be therapeutically halted or reversed. In this review, we highlight the current progress in this field with respect to numerous pathological factors and discuss the possible mechanisms. Moreover, these new observations also show that ATI can be mechanistically reversed.
Collapse
Affiliation(s)
- Abdur Rahman Ansari
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China.,Section of Anatomy and Histology, Department of Basic Sciences, College of Veterinary and Animal Sciences (CVAS), Jhang, Pakistan.,University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan
| | - Huazhen Liu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China.
| |
Collapse
|
33
|
Moysi E, Estes JD, Petrovas C. Novel Imaging Methods for Analysis of Tissue Resident Cells in HIV/SIV. Curr HIV/AIDS Rep 2016; 13:38-43. [PMID: 26830285 DOI: 10.1007/s11904-016-0300-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of advanced tissue-imaging methodologies has greatly facilitated the study of molecular mechanisms and cellular interactions in humans and animal models of disease. Particularly, in HIV research, there is an ever-increasing demand for a comprehensive analysis of immune cell dynamics at tissue level stemming from the need to advance our understanding of those interactions that regulate the generation of adaptive antigen-specific immune responses. The latter is critical for the development of vaccines to elicit broadly neutralizing antibodies as well as for the discovery of novel targets for immuno-therapies to strengthen the cytolytic arm of the immune system at local level. In this review, we focus on current and emerging imaging technologies, discuss their strengths and limitations, and examine how such technologies can inform the development of new treatments and vaccination strategies. We also present some perspective on the future of the technology development.
Collapse
Affiliation(s)
- Eirini Moysi
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136-1013, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Constantinos Petrovas
- Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health, Building 40, 40 Convent Drive, Bethesda, MD, 20892-3005, USA.
| |
Collapse
|
34
|
Hong JJ, Chang KT, Villinger F. The Dynamics of T and B Cells in Lymph Node during Chronic HIV Infection: TFH and HIV, Unhappy Dance Partners? Front Immunol 2016; 7:522. [PMID: 27920778 PMCID: PMC5118424 DOI: 10.3389/fimmu.2016.00522] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/09/2016] [Indexed: 11/13/2022] Open
Abstract
Although the dynamics of germinal center (GC) formation, follicular helper T (TFH) cell recruitment to B cell follicles within lymphoid organs, and changes of lymphoid tissue architecture in HIV/SIV infection have been documented, the underlying immunopathology remains unclear. Here, we summarize what is known regarding the kinetics of TFH cells and GC B cells during the course of infection as well as the potential immunopathological features associated with structural changes in the lymphoid compartment. This review also explores the implications of cell dynamics in the formation and maintenance of viral reservoirs in hyperplastic follicles of secondary lymphoid organs before and after viral suppressive antiretroviral therapy.
Collapse
Affiliation(s)
- Jung Joo Hong
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB) , Cheongju , South Korea
| | - Kyu-Tae Chang
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB) , Cheongju , South Korea
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana Lafayette , Lafayette, LA , USA
| |
Collapse
|
35
|
The first 24 h: targeting the window of opportunity for antibody-mediated protection against HIV-1 transmission. Curr Opin HIV AIDS 2016; 11:561-568. [PMID: 27559708 DOI: 10.1097/coh.0000000000000319] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW I will review evidence that antibodies protect against HIV-1 transmission in a short window of opportunity, involving neutralization, Fc-mediated effector function, or both. RECENT FINDINGS The last decade witnessed a dramatic progress in the understanding of antibody-mediated protection against HIV-1, including active and passive immunization studies in nonhuman primates; association between reduced infection risk and the specificities and function of antibodies in the RV144 clinical trial; identification of potent, broadly neutralizing antibodies; high-resolution structural studies of the HIV-1 envelope trimer; and an increasing appreciation that Fc-mediated effector function is critical to protection against transmission for neutralizing and nonneutralizing antibodies. Less information is known about how antibodies protect in situ, except that they must do in the first 24 h after exposure. New evidence suggests that antibodies protect in an acute innate immune environment involving the NXLRX1 inflammasome and transforming growth factor beta (TGF-β) that favors infection and rapid dissemination of CCR6RORγ Th17 cells. SUMMARY These recent findings set the stage for understanding how antibodies can prevent the transmission of HIV-1. In this context, antibodies must prevent infection in an innate immune environment that strongly favors transmission. This information is key for the development of a vaccine against HIV-1.
Collapse
|
36
|
Wang X, Ziani W, Xu H. Changes in Follicular CD4+ T Helper Cells as a Marker for Evaluating Disease Progression in the Competition between HIV and Host Immunity. Front Immunol 2016; 7:474. [PMID: 27843442 PMCID: PMC5087249 DOI: 10.3389/fimmu.2016.00474] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/19/2016] [Indexed: 11/17/2022] Open
Abstract
Follicular CD4+ T helper (TFH) cells interact with B cells in follicular germinal centers and play a prominent role in promoting effective humoral immune responses to pathogens, providing help for B cell development and antibody affinity maturation. Recent studies indicate TFH cells are expanded in HIV/SIV chronic infection, or depleted in terminal stages of disease, yet relatively maintained in elite controllers when compared with uninfected controls. A better understanding of the mechanisms behind these immunologic abnormalities may lead to more effective vaccination and therapeutic strategies. Here, we review recent findings of TFH cells in HIV/SIV infection and discuss the correlation of changes and function of TFH cells with host immunity. Dysregulation or depletion of CD4+ TFH cells likely plays a major role in the inability of HIV-infected patients to mount effective immune responses.
Collapse
Affiliation(s)
- Xiaolei Wang
- Tulane National Primate Research Center, Tulane University School of Medicine , Covington, LA , USA
| | - Widade Ziani
- Tulane National Primate Research Center, Tulane University School of Medicine , Covington, LA , USA
| | - Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine , Covington, LA , USA
| |
Collapse
|
37
|
Bouteloup V, Sabin C, Mocroft A, Gras L, Pantazis N, Le Moing V, d'Arminio Monforte A, Mary-Krause M, Roca B, Miro JM, Battegay M, Brockmeyer N, Berenguer J, Morlat P, Obel N, De Wit S, Fätkenheuer G, Zangerle R, Ghosn J, Pérez-Hoyos S, Campbell M, Prins M, Chêne G, Meyer L, Dorrucci M, Torti C, Thiébaut R. Reference curves for CD4 T-cell count response to combination antiretroviral therapy in HIV-1-infected treatment-naïve patients. HIV Med 2016; 18:33-44. [PMID: 27625009 DOI: 10.1111/hiv.12389] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2016] [Indexed: 12/01/2022]
Abstract
OBJECTIVES The aim of this work was to provide a reference for the CD4 T-cell count response in the early months after the initiation of combination antiretroviral therapy (cART) in HIV-1-infected patients. METHODS All patients in the Collaboration of Observational HIV Epidemiological Research Europe (COHERE) cohort who were aged ≥ 18 years and started cART for the first time between 1 January 2005 and 1 January 2010 and who had at least one available measurement of CD4 count and a viral load ≤ 50 HIV-1 RNA copies/mL at 6 months (± 3 months) after cART initiation were included in the study. Unadjusted and adjusted references curves and predictions were obtained using quantile regressions. RESULTS A total of 28 992 patients were included in the study. The median CD4 T-cell count at treatment initiation was 249 [interquartile range (IQR) 150, 336] cells/μL. The median observed CD4 counts at 6, 9 and 12 months were 382 (IQR 256, 515), 402 (IQR 274, 543) and 420 (IQR 293, 565) cells/μL. The two main factors explaining the variation of CD4 count at 6 months were AIDS stage and CD4 count at cART initiation. A CD4 count increase of ≥ 100 cells/mL is generally required in order that patients stay 'on track' (i.e. with a CD4 count at the same percentile as when they started), with slightly higher gains required for those starting with CD4 counts in the higher percentiles. Individual predictions adjusted for factors influencing CD4 count were more precise. CONCLUSIONS Reference curves aid the evaluation of the immune response early after antiretroviral therapy initiation that leads to viral control.
Collapse
Affiliation(s)
- V Bouteloup
- CIC 1401, CHU de Bordeaux, Bordeaux, France.,INSERM U1219 - Centre Inserm Bordeaux Population Health, Université de Bordeaux, Bordeaux, France.,ISPED, Centre INSERM U1219-Bordeaux Population Health, Université de Bordeaux, Bordeaux, France
| | - C Sabin
- Research Department of Infection & Population Health, UCL, London, UK
| | - A Mocroft
- Research Department of Infection & Population Health, UCL, London, UK
| | - L Gras
- Stichting HIV Monitoring, Amsterdam, The Netherlands
| | - N Pantazis
- Department of Hygiene, Epidemiology & Medical Statistics, Athens University Medical School, Athens, Greece
| | - V Le Moing
- Montpellier University, Montpellier, France
| | - A d'Arminio Monforte
- Infectious Diseases Unit, Department of Health Sciences, San Paolo University Hospital, Milan, Italy
| | - M Mary-Krause
- INSERM, Institut Pierre Louis d'épidémiologie et de Santé Publique (IPLESP UMRS 1136), UPMC Univ Paris 06, Sorbonne Universités, F-75013, Paris, France
| | - B Roca
- Hospital General of Castellon, Castellón, Spain
| | - J M Miro
- Infectious Diseases Service. Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - M Battegay
- Division of Infectious Diseases and Hospital Epidemiology, Department of Clinical Research, University Hospital of Basel, Basel, Switzerland
| | - N Brockmeyer
- Department of Dermatology, Venerology - Center for Sexual Health and Medicine, Ruhr-Universität Bochum, Bochum, Germany
| | - J Berenguer
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - P Morlat
- INSERM U1219 - Centre Inserm Bordeaux Population Health, Université de Bordeaux, Bordeaux, France.,Service de Médecine Interne et Maladies Infectieuses, Hôpital Saint-André, Bordeaux, France
| | - N Obel
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - S De Wit
- Department of Infectious Diseases, St Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - G Fätkenheuer
- Department of Internal Medicine, University of Cologne and German Centre for Infection Research (DZIF), Cologne, Germany
| | - R Zangerle
- Medical University Innsbruck, Innsbruck, Austria
| | - J Ghosn
- APHP, Unité Fonctionnelle de Thérapeutique en Immuno-Infectiologie, Centre Hospitalier Universitaire Hôtel Dieu, Paris, France.,Faculté de Médecine Site Necker, Sorbonne Paris Cité, Université Paris Descartes, EA 7327, Paris, France
| | - S Pérez-Hoyos
- Vall d'Hebrón Institut de Recerca (VHIR), Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M Campbell
- CHIP, Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - M Prins
- Division of Infectious Diseases, Department of Internal Medicine, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands.,Department of Infectious Diseases, Public Health Service, Amsterdam, The Netherlands
| | - G Chêne
- CIC 1401, CHU de Bordeaux, Bordeaux, France.,INSERM U1219 - Centre Inserm Bordeaux Population Health, Université de Bordeaux, Bordeaux, France.,ISPED, Centre INSERM U1219-Bordeaux Population Health, Université de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Pole de Sante Publique, Service d'Information Medicale, F-33000, Bordeaux, France
| | - L Meyer
- INSERM, U1018, Epidemiology of HIV, Reproduction, Paediatrics, CESP; University Paris-Sud, Paris, France.,Department of Public Health and Epidemiology, Bicêtre Hospital, AP-HP, Le Kremlin Bicêtre, Paris, France
| | - M Dorrucci
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - C Torti
- Unit of Infectious and Tropical Diseases, Department of Medical and Surgical Sciences, University "Magna Graecia", Catanzaro, Italy
| | - R Thiébaut
- INSERM U1219 - Centre Inserm Bordeaux Population Health, Université de Bordeaux, Bordeaux, France.,ISPED, Centre INSERM U1219-Bordeaux Population Health, Université de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Pole de Sante Publique, Service d'Information Medicale, F-33000, Bordeaux, France
| | | |
Collapse
|
38
|
Koroleva EP, Fu YX, Tumanov AV. Lymphotoxin in physiology of lymphoid tissues - Implication for antiviral defense. Cytokine 2016; 101:39-47. [PMID: 27623349 DOI: 10.1016/j.cyto.2016.08.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022]
Abstract
Lymphotoxin (LT) is a member of the tumor necrosis factor (TNF) superfamily of cytokines which serves multiple functions, including the control of lymphoid organ development and maintenance, as well as regulation of inflammation and autoimmunity. Although the role of LT in organogenesis and maintenance of lymphoid organs is well established, the contribution of LT pathway to homeostasis of lymphoid organs during the immune response to pathogens is less understood. In this review, we highlight recent advances on the role of LT pathway in antiviral immune responses. We discuss the role of LT signaling in lymphoid organ integrity, type I IFN production and regulation of protection and immunopathology during viral infections. We further discuss the potential of therapeutic targeting LT pathway for controlling immunopathology and antiviral protection.
Collapse
Affiliation(s)
- Ekaterina P Koroleva
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas School of Medicine, UT Health Science Center, San Antonio, TX, USA; Trudeau Institute, Saranac Lake, NY
| | - Yang-Xin Fu
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexei V Tumanov
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas School of Medicine, UT Health Science Center, San Antonio, TX, USA; Trudeau Institute, Saranac Lake, NY.
| |
Collapse
|
39
|
Nazari B, Rice LM, Stifano G, Barron AMS, Wang YM, Korndorf T, Lee J, Bhawan J, Lafyatis R, Browning JL. Altered Dermal Fibroblasts in Systemic Sclerosis Display Podoplanin and CD90. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2650-64. [PMID: 27565038 DOI: 10.1016/j.ajpath.2016.06.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 05/02/2016] [Accepted: 06/03/2016] [Indexed: 12/21/2022]
Abstract
Tissue injury triggers the activation and differentiation of multiple cell types to minimize damage and initiate repair processes. In systemic sclerosis, these repair processes appear to run unchecked, leading to aberrant remodeling and fibrosis of the skin and multiple internal organs, yet the fundamental pathological defect remains unknown. We describe herein a transition wherein the abundant CD34(+) dermal fibroblasts present in healthy human skin disappear in the skin of systemic sclerosis patients, and CD34(-), podoplanin(+), and CD90(+) fibroblasts appear. This transition is limited to the upper dermis in several inflammatory skin diseases, yet in systemic sclerosis, it can occur in all regions of the dermis. In vitro, primary dermal fibroblasts readily express podoplanin in response to the inflammatory stimuli tumor necrosis factor and IL-1β. Furthermore, we show that on acute skin injury in both human and murine settings, this transition occurs quickly, consistent with a response to inflammatory signaling. Transitioned fibroblasts partially resemble the cells that form the reticular networks in organized lymphoid tissues, potentially linking two areas of fibroblast research. These results allow for the visualization and quantification of a basic stage of fibroblast differentiation in inflammatory and fibrotic diseases in the skin.
Collapse
Affiliation(s)
- Banafsheh Nazari
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts
| | - Lisa M Rice
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts
| | - Giuseppina Stifano
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts
| | - Alexander M S Barron
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts; Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts
| | - Yu Mei Wang
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts
| | - Tess Korndorf
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts
| | - Jungeun Lee
- Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts
| | - Robert Lafyatis
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts; Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jeffrey L Browning
- Section of Rheumatology, Boston University School of Medicine, Boston, Massachusetts; Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
40
|
Deleage C, Turkbey B, Estes JD. Imaging lymphoid tissues in nonhuman primates to understand SIV pathogenesis and persistence. Curr Opin Virol 2016; 19:77-84. [PMID: 27490446 PMCID: PMC5021606 DOI: 10.1016/j.coviro.2016.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/01/2016] [Accepted: 07/03/2016] [Indexed: 02/04/2023]
Abstract
CD4+ T cells are the primary HIV-1 target cell, with the vast majority of these cells residing within lymphoid tissue compartments throughout the body. Predictably, HIV-1 infection, replication, localization, reservoir establishment and persistence, as well as associated host immune and inflammatory responses and disease pathology principally take place within the tissues of the immune system. By virture of the fact that the virus-host struggle is played out within lymphoid and additional tissues compartments in HIV-1 infected individuals it is critical to understand HIV-1 infection and disease within these relevant tissue sites; however, there are obvious limitations to studying these dynamic processes in humans. Nonhuman primate (NHP) research has provided a vital bridge between basic and preclinical research and clinical studies, with experimental SIV infection of NHP models offering unique opportunities to understand key processes of HIV-1 infection and disease that are either not practically feasible or ethical in HIV-1 infected humans. In this review we will discuss current approaches to studying the tissue based immunopathogenesis of AIDS virus infection in NHPs, including both analyses of tissues obtained at biopsy or necropsy and complementary non-invasive imaging approaches that may have practical utility in monitoring HIV-1 disease in the clinical setting.
Collapse
Affiliation(s)
- Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, MD 21702, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, MD 20814, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, MD 21702, USA.
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW The introduction of effective antiretroviral therapy (ART) has transformed HIV infection from a deadly to a chronic infection. Despite its successes in reducing mortality, ART fails to cure HIV allowing HIV to persist in vivo. HIV persistence under ART is thought to be mediated by a combination of latent infection of long-lived cells, homeostatic proliferation of latently infected cells, anatomic sanctuaries, and low-level virus replication. To understand the contribution of specific cell types and anatomic sites to virus persistence in vivo animal models are necessary. RECENT FINDINGS The advancements in ART and our understanding of animal models have facilitated the development of models of HIV persistence in nonhuman primates and mice. Simian immunodeficiency virus (SIV) or simian/HIV infection (SHIV) of rhesus and pigtail macaques followed by effective ART represents the most faithful animal model of HIV persistence. HIV infection of humanized mice also provides a useful model for answering specific questions regarding virus persistence in a uniquely mutable system. SUMMARY In this review, we describe the most recent findings using animal models of HIV persistence. We will first describe the important aspects of HIV infection that SIV/SHIV infection of nonhuman primates are able to recapitulate, then we will discuss some recent studies that have used these models to understand viral persistence.
Collapse
|
42
|
Lymphoid Tissue Mesenchymal Stromal Cells in Development and Tissue Remodeling. Stem Cells Int 2016; 2016:8419104. [PMID: 27190524 PMCID: PMC4846763 DOI: 10.1155/2016/8419104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/20/2016] [Indexed: 12/28/2022] Open
Abstract
Secondary lymphoid organs (SLOs) are sites that facilitate cell-cell interactions required for generating adaptive immune responses. Nonhematopoietic mesenchymal stromal cells have been shown to play a critical role in SLO function, organization, and tissue homeostasis. The stromal microenvironment undergoes profound remodeling to support immune responses. However, chronic inflammatory conditions can promote uncontrolled stromal cell activation and aberrant tissue remodeling including fibrosis, thus leading to tissue damage. Despite recent advancements, the origin and role of mesenchymal stromal cells involved in SLO development and remodeling remain unclear.
Collapse
|
43
|
Abstract
OBJECTIVES AIDS is caused by CD4 T-cell depletion. Although combination antiretroviral therapy can restore blood T-cell numbers, the clonal diversity of the reconstituting cells, critical for immunocompetence, is not well defined. METHODS We performed an extensive analysis of parameters of thymic function in perinatally HIV-1-infected (n = 39) and control (n = 28) participants ranging from 13 to 23 years of age. CD4 T cells including naive (CD27 CD45RA) and recent thymic emigrant (RTE) (CD31/CD45RA) cells, were quantified by flow cytometry. Deep sequencing was used to examine T-cell receptor (TCR) sequence diversity in sorted RTE CD4 T cells. RESULTS Infected participants had reduced CD4 T-cell levels with predominant depletion of the memory subset and preservation of naive cells. RTE CD4 T-cell levels were normal in most infected individuals, and enhanced thymopoiesis was indicated by higher proportions of CD4 T cells containing TCR recombination excision circles. Memory CD4 T-cell depletion was highly associated with CD8 T-cell activation in HIV-1-infected persons and plasma interlekin-7 levels were correlated with naive CD4 T cells, suggesting activation-driven loss and compensatory enhancement of thymopoiesis. Deep sequencing of CD4 T-cell receptor sequences in well compensated infected persons demonstrated supranormal diversity, providing additional evidence of enhanced thymic output. CONCLUSION Despite up to two decades of infection, many individuals have remarkable thymic reserve to compensate for ongoing CD4 T-cell loss, although there is ongoing viral replication and immune activation despite combination antiretroviral therapy. The longer term sustainability of this physiology remains to be determined.
Collapse
|
44
|
Abstract
Although the replicative life cycle of HIV within CD4 T cells is understood in molecular detail, less is known about how this human retrovirus promotes the loss of CD4 T lymphocytes. It is this cell death process that drives clinical progression to acquired immune deficiency syndrome (AIDS). Recent studies have highlighted how abortive infection of resting and thus nonpermissive CD4 T cells in lymphoid tissues triggers a lethal innate immune response against the incomplete DNA products generated by inefficient viral reverse transcription in these cells. Sensing of these DNA fragments results in pyroptosis, a highly inflammatory form of programmed cell death, that potentially further perpetuates chronic inflammation and immune activation. As discussed here, these studies cast CD4 T cell death during HIV infection in a different light. Further, they identify drug targets that may be exploited to both block CD4 T cell demise and the chronic inflammatory response generated during pyroptosis.
Collapse
Affiliation(s)
- Gilad Doitsh
- Gladstone Institute of Virology and Immunology, 1650 Owens Street, San Francisco, CA 94158, USA.
| | - Warner C Greene
- Gladstone Institute of Virology and Immunology, 1650 Owens Street, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
45
|
Silva SL, Sousa AE. Establishment and Maintenance of the Human Naïve CD4 + T-Cell Compartment. Front Pediatr 2016; 4:119. [PMID: 27843891 PMCID: PMC5086629 DOI: 10.3389/fped.2016.00119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/17/2016] [Indexed: 12/26/2022] Open
Abstract
The naïve CD4+ T-cell compartment is considered essential to guarantee immune competence throughout life. Its replenishment with naïve cells with broad diverse receptor repertoire, albeit with reduced self-reactivity, is ensured by the thymus. Nevertheless, cumulative data support a major requirement of post-thymic proliferation both for the establishment of the human peripheral naïve compartment during the accelerated somatic growth of childhood, as well as for its lifelong maintenance. Additionally, a dynamic equilibrium is operating at the cell level to fine-tune the T-cell receptor threshold to activation and survival cues, in order to counteract the continuous naïve cell loss by death or conversion into memory/effector cells. The main players in these processes are low-affinity self-peptide/MHC and cytokines, particularly IL-7. Moreover, although naïve CD4+ T-cells are usually seen as a homogeneous population regarding stage of maturation and cell differentiation, increasing evidence points to a variety of phenotypic and functional subsets with distinct homeostatic requirements. The paradigm of cells committed to a distinct lineage in the thymus are the naïve regulatory T-cells, but other functional subpopulations have been identified based on their time span after thymic egress, phenotypic markers, such as CD31, or cytokine production, namely IL-8. Understanding the regulation of these processes is of utmost importance to promote immune reconstitution in several clinical settings, namely transplantation, persistent infections, and aging. In this mini review, we provide an overview of the mechanisms underlying human naïve CD4+ T-cell homeostasis, combining clinical data, experimental studies, and modeling approaches.
Collapse
Affiliation(s)
- Susana L Silva
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Centro de Imunodeficiências Primárias, Lisboa, Portugal; Clinica Universitária de Imunoalergologia, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisboa, Portugal
| | - Ana E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Centro de Imunodeficiências Primárias, Lisboa, Portugal
| |
Collapse
|
46
|
Wang S, Hottz P, Schechter M, Rong L. Modeling the Slow CD4+ T Cell Decline in HIV-Infected Individuals. PLoS Comput Biol 2015; 11:e1004665. [PMID: 26709961 PMCID: PMC4692447 DOI: 10.1371/journal.pcbi.1004665] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023] Open
Abstract
The progressive loss of CD4+ T cell population is the hallmark of HIV-1 infection but the mechanism underlying the slow T cell decline remains unclear. Some recent studies suggested that pyroptosis, a form of programmed cell death triggered during abortive HIV infection, is associated with the release of inflammatory cytokines, which can attract more CD4+ T cells to be infected. In this paper, we developed mathematical models to study whether this mechanism can explain the time scale of CD4+ T cell decline during HIV infection. Simulations of the models showed that cytokine induced T cell movement can explain the very slow decline of CD4+ T cells within untreated patients. The long-term CD4+ T cell dynamics predicted by the models were shown to be consistent with available data from patients in Rio de Janeiro, Brazil. Highly active antiretroviral therapy has the potential to restore the CD4+ T cell population but CD4+ response depends on the effectiveness of the therapy, when the therapy is initiated, and whether there are drug sanctuary sites. The model also showed that chronic inflammation induced by pyroptosis may facilitate persistence of the HIV latent reservoir by promoting homeostatic proliferation of memory CD4+ cells. These results improve our understanding of the long-term T cell dynamics in HIV-1 infection, and support that new treatment strategies, such as the use of caspase-1 inhibitors that inhibit pyroptosis, may maintain the CD4+ T cell population and reduce the latent reservoir size. The CD4+ T cell population within HIV-infected individuals declines slowly as disease progresses. When CD4+ cells drop to below 200 cells/ul, the infection is usually considered to enter the late stage, i.e., acquired immune deficiency syndrome (AIDS). CD4+ T cell depletion can take many years but the biological events underlying such slow decline are not well understood. Some studies showed that the majority of infected T cells in lymph nodes die by pyroptosis, a form of programmed cell death, which can release inflammatory signals attracting more CD4+ T cells to be infected. We developed mathematical models to describe this process and explored whether they can generate the long-term CD4+ T cell decline. We showed that pyroptosis induced cell movement can explain the slow time scale of CD4+ T cell depletion and that pyroptosis may also contribute to the persistence of latently infected cells, which represent a major obstacle to HIV eradication. The modeling prediction agrees with patient data in Rio de Janeiro, Brazil. These results suggest that a combination of current treatment regimens and caspase-1 inhibitor that can inhibit pyroptosis might provide a new way to maintain the CD4+ T cell population and eradicate the HIV latent reservoir.
Collapse
Affiliation(s)
- Sunpeng Wang
- Department of Biology, New York University, New York, New York, United States of America
| | - Patricia Hottz
- Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauro Schechter
- Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Projeto Praça Onze, Hospital Escola São Francisco de Assis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Libin Rong
- Department of Mathematics and Statistics, and Center for Biomedical Research, Oakland University, Rochester, Michigan, United States of America
- * E-mail:
| |
Collapse
|
47
|
Vergnon-Miszczycha D, Lucht F, Roblin X, Pozzetto B, Paul S, Bourlet T. [Key role played by the gut associated lymphoid tissue during human immunodeficiency virus infection]. Med Sci (Paris) 2015; 31:1092-101. [PMID: 26672662 DOI: 10.1051/medsci/20153112012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The gut associated lymphoid tissue (GALT) is the site of numerous immunological disturbances during HIV-1 infection. It constitutes the largest reservoir for HIV, not or very poorly susceptible to antiretroviral therapy (ART), making it a major obstacle to HIV cure. Moreover, the GALT is involved in systemic immune activation in HIV-infected individuals: intestinal damage due to viral replication and severe CD4(+) T cell depletion in the GALT leads to microbial translocation, a key driver of immune activation, and in turn, disease progression. In this review, we describe the role of the GALT in HIV infection and we discuss therapeutic options to decrease the intestinal viral reservoir and to preserve immune function in the gut of HIV-infected people. Achieving these goals is necessary for a long-term infection control after the interruption of ART.
Collapse
Affiliation(s)
- Delphine Vergnon-Miszczycha
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Service de maladies infectieuses et tropicales, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Frédéric Lucht
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Service de maladies infectieuses et tropicales, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Xavier Roblin
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Service de gastro-entérologie, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Bruno Pozzetto
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Laboratoire des agents infectieux et d'hygiène, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Stéphane Paul
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Laboratoire d'immunologie, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Thomas Bourlet
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Laboratoire des agents infectieux et d'hygiène, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| |
Collapse
|
48
|
Song E, Seo H, Choe K, Hwang Y, Ahn J, Ahn S, Kim P. Optical clearing based cellular-level 3D visualization of intact lymph node cortex. BIOMEDICAL OPTICS EXPRESS 2015; 6:4154-64. [PMID: 26504662 PMCID: PMC4605071 DOI: 10.1364/boe.6.004154] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 05/03/2023]
Abstract
Lymph node (LN) is an important immune organ that controls adaptive immune responses against foreign pathogens and abnormal cells. To facilitate efficient immune function, LN has highly organized 3D cellular structures, vascular and lymphatic system. Unfortunately, conventional histological analysis relying on thin-sliced tissue has limitations in 3D cellular analysis due to structural disruption and tissue loss in the processes of fixation and tissue slicing. Optical sectioning confocal microscopy has been utilized to analyze 3D structure of intact LN tissue without physical tissue slicing. However, light scattering within biological tissues limits the imaging depth only to superficial portion of LN cortex. Recently, optical clearing techniques have shown enhancement of imaging depth in various biological tissues, but their efficacy for LN are remained to be investigated. In this work, we established optical clearing procedure for LN and achieved 3D volumetric visualization of the whole cortex of LN. More than 4 times improvement in imaging depth was confirmed by using LN obtained from H2B-GFP/actin-DsRed double reporter transgenic mouse. With adoptive transfer of GFP expressing B cells and DsRed expressing T cells and fluorescent vascular labeling by anti-CD31 and anti-LYVE-1 antibody conjugates, we successfully visualized major cellular-level structures such as T-cell zone, B-cell follicle and germinal center. Further, we visualized the GFP expressing metastatic melanoma cell colony, vasculature and lymphatic vessels in the LN cortex.
Collapse
|
49
|
Xu H, Wang X, Malam N, Lackner AA, Veazey RS. Persistent Simian Immunodeficiency Virus Infection Causes Ultimate Depletion of Follicular Th Cells in AIDS. THE JOURNAL OF IMMUNOLOGY 2015; 195:4351-7. [PMID: 26408660 DOI: 10.4049/jimmunol.1501273] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023]
Abstract
CD4(+) T follicular helper (Tfh) cells are critical for the generation of humoral immune responses to pathogenic infections, providing help for B cell development, survival, and affinity maturation of Abs. Although CD4(+) Tfh cells are reported to accumulate in HIV or SIV infection, we found that germinal center Tfh cells, defined in this study as CXCR5(+)PD-1(HIGH)CD4(+) T cells, did not consistently accumulate in chronically SIV-infected rhesus macaques compared with those infected with less pathogenic simian HIV, vaccinated and SIVmac-challenged, or SIVmac-infected Mamu-A*01(+) macaques, all of which are associated with some control of virus replication and slower disease progression. Interestingly, CXCR5(+)PD-1(HIGH) Tfh cells in lymphoid tissues were eventually depleted in macaques with AIDS compared with the other cohorts. Chronic activation and proliferation of CXCR5(+)PD-1(HIGH) Tfh were increased, but PD-L2 expression was downregulated on B cells, possibly resulting in germinal center Tfh cell apoptosis. Together, these findings suggest that changes in CXCR5(+)PD-1(HIGH) Tfh cells in lymph nodes correlate with immune control during infection, and their loss or dysregulation contribute to impairment of B cell responses and progression to AIDS.
Collapse
Affiliation(s)
- Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Xiaolei Wang
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Naomi Malam
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Andrew A Lackner
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Ronald S Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| |
Collapse
|
50
|
Lo YR, Chu C, Ananworanich J, Excler JL, Tucker JD. Stakeholder Engagement in HIV Cure Research: Lessons Learned from Other HIV Interventions and the Way Forward. AIDS Patient Care STDS 2015; 29:389-99. [PMID: 26061668 DOI: 10.1089/apc.2014.0348] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Clinical and basic science advances have raised considerable hope for achieving an HIV cure by accelerating research. This research is dominated primarily by issues about the nature and design of current and future clinical trials. Stakeholder engagement for HIV cure remains in its early stages. Our analysis examines timing and mechanisms of historical stakeholder engagement in other HIV research areas for HIV-uninfected individuals [vaccine development and pre-exposure prophylaxis (PrEP)], and HIV-infected individuals (treatment as prevention, prevention of mother-to-child transmission, and treatment of acute HIV infection) and articulate a plan for HIV cure stakeholder engagement. The experience from HIV vaccine development shows that early engagement of stakeholders helped manage expectations, mitigating the failure of several vaccine trials, while paving the way for subsequent trials. The relatively late engagement of HIV stakeholders in PrEP research may partly explain some of the implementation challenges. The treatment-related stakeholder engagement was strong and community-led from the onset and helped translation from research to implementation. We outline five steps to initiate and sustain stakeholder engagement in HIV cure research and conclude that stakeholder engagement represents a key investment in which stakeholders mutually agree to share knowledge, benefits, and risk of failure. Effective stakeholder engagement prevents misconceptions. As HIV cure research advances from early trials involving subjects with generally favorable prognosis to studies involving greater risk and uncertainty, success may depend on early and deliberate engagement of stakeholders.
Collapse
Affiliation(s)
- Ying-Ru Lo
- HIV, Hepatitis and STI Unit, World Health Organization, Regional Office for the Western Pacific, Manila, The Philippines
| | - Carissa Chu
- University of California San Francisco School of Medicine, San Francisco, California
- University of North Carolina Project-China, Guangzhou, P.R. China
| | - Jintanat Ananworanich
- US Military HIV Research Program, Bethesda, Maryland
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland
| | - Jean-Louis Excler
- US Military HIV Research Program, Bethesda, Maryland
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland
| | - Joseph D. Tucker
- University of North Carolina Project-China, Guangzhou, P.R. China
- Institute of Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|