151
|
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
|
152
|
Programed death-1/programed death-ligand 1 expression in lymph nodes of HIV infected patients: results of a pilot safety study in rhesus macaques using anti-programed death-ligand 1 (Avelumab). AIDS 2016; 30:2487-2493. [PMID: 27490642 PMCID: PMC5051527 DOI: 10.1097/qad.0000000000001217] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The programed death-1 (PD1)/programed death-ligand 1 (PD-L1) pathway plays a critical role in balancing immunity and host immunopathology. During chronic HIV/SIV infection, there is persistent immune activation accompanied by accumulation of virus-specific cells with terminally differentiated phenotypes and expression of regulatory receptors such as PD1. These observations led us to hypothesize that the PD1/PD-L1 pathway contributes to the functional dysregulation and ineffective viral control, and its blockade may be a potential immunotherapeutic target.
Collapse
|
153
|
Thompson M, Heath SL, Sweeton B, Williams K, Cunningham P, Keele BF, Sen S, Palmer BE, Chomont N, Xu Y, Basu R, Hellerstein MS, Kwa S, Robinson HL. DNA/MVA Vaccination of HIV-1 Infected Participants with Viral Suppression on Antiretroviral Therapy, followed by Treatment Interruption: Elicitation of Immune Responses without Control of Re-Emergent Virus. PLoS One 2016; 11:e0163164. [PMID: 27711228 PMCID: PMC5053438 DOI: 10.1371/journal.pone.0163164] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/14/2016] [Indexed: 12/01/2022] Open
Abstract
GV-TH-01, a Phase 1 open-label trial of a DNA prime—Modified Vaccinia Ankara (MVA) boost vaccine (GOVX-B11), was undertaken in HIV infected participants on antiretroviral treatment (ART) to evaluate safety and vaccine-elicited T cell responses, and explore the ability of elicited CD8+ T cells to control viral rebound during analytical treatment interruption (TI). Nine men who began antiretroviral therapy (ART) within 18 months of seroconversion and had sustained plasma HIV-1 RNA <50 copies/mL for at least 6 months were enrolled. Median age was 38 years, median pre-ART HIV-1 RNA was 140,000 copies/ml and mean baseline CD4 count was 755/μl. Two DNA, followed by 2 MVA, inoculations were given 8 weeks apart. Eight subjects completed all vaccinations and TI. Clinical and laboratory adverse events were generally mild, with no serious or grade 4 events. Only reactogenicity events were considered related to study drug. No treatment emergent viral resistance was seen. The vaccinations did not reduce viral reservoirs and virus re-emerged in all participants during TI, with a median time to re-emergence of 4 weeks. Eight of 9 participants had CD8+ T cells that could be stimulated by vaccine-matched Gag peptides prior to vaccination. Vaccinations boosted these responses as well as eliciting previously undetected CD8+ responses. Elicited T cells did not display signs of exhaustion. During TI, temporal patterns of viral re-emergence and Gag-specific CD8+ T cell expansion suggested that vaccine-specific CD8+ T cells had been stimulated by re-emergent virus in only 2 of 8 participants. In these 2, transient decreases in viremia were associated with Gag selection in known CD8+ T cell epitopes. We hypothesize that escape mutations, already archived in the viral reservoir, plus a poor ability of CD8+ T cells to traffic to and control virus at sites of re-emergence, limited the therapeutic efficacy of the DNA/MVA vaccine. TRIAL REGISTRATION clinicaltrials.gov NCT01378156.
Collapse
Affiliation(s)
- Melanie Thompson
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Sonya L. Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Bentley Sweeton
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Kathy Williams
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Pamela Cunningham
- Alabama Vaccine Research Clinic, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sharon Sen
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Brent E. Palmer
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Nicolas Chomont
- Centre de recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Canada
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Rahul Basu
- GeoVax, Inc., Atlanta, Georgia, United States of America
| | | | - Suefen Kwa
- GeoVax, Inc., Atlanta, Georgia, United States of America
| | | |
Collapse
|
154
|
Miles B, Miller SM, Folkvord JM, Levy DN, Rakasz EG, Skinner PJ, Connick E. Follicular Regulatory CD8 T Cells Impair the Germinal Center Response in SIV and Ex Vivo HIV Infection. PLoS Pathog 2016; 12:e1005924. [PMID: 27716848 PMCID: PMC5055335 DOI: 10.1371/journal.ppat.1005924] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/10/2016] [Indexed: 12/21/2022] Open
Abstract
During chronic HIV infection, viral replication is concentrated in secondary lymphoid follicles. Cytotoxic CD8 T cells control HIV replication in extrafollicular regions, but not in the follicle. Here, we show CXCR5hiCD44hiCD8 T cells are a regulatory subset differing from conventional CD8 T cells, and constitute the majority of CD8 T cells in the follicle. This subset, CD8 follicular regulatory T cells (CD8 TFR), expand in chronic SIV infection, exhibit enhanced expression of Tim-3 and IL-10, and express less perforin compared to conventional CD8 T cells. CD8 TFR modestly limit HIV replication in follicular helper T cells (TFH), impair TFH IL-21 production via Tim-3, and inhibit IgG production by B cells during ex vivo HIV infection. CD8 TFR induce TFH apoptosis through HLA-E, but induce less apoptosis than conventional CD8 T cells. These data demonstrate that a unique regulatory CD8 population exists in follicles that impairs GC function in HIV infection.
Collapse
Affiliation(s)
- Brodie Miles
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver, Colorado, United States of America
| | - Shannon M. Miller
- Department of Immunology and Microbiology, School of Medicine, Anschutz Medical Campus, University of Colorado Denver, Colorado, United States of America
| | - Joy M. Folkvord
- Division of Infectious Diseases, University of Arizona, Arizona, United States of America
| | - David N. Levy
- Department of Basic Science, New York University College of Dentistry, New York, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minnesota, United States of America
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Arizona, United States of America
| |
Collapse
|
155
|
Does persistent HIV replication explain continued lymphoma incidence in the era of effective antiretroviral therapy? Curr Opin Virol 2016; 20:71-77. [PMID: 27665065 DOI: 10.1016/j.coviro.2016.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/16/2022]
Abstract
Non-Hodgkin lymphomas are highly increased in incidence in individuals infected with HIV, and this continues to be the case in spite of highly effective combined antiretroviral therapy (cART). New evidence has demonstrated that while successful virtual recovery of CD4 counts and elimination of HIV from peripheral blood can be achieved with cART, viral replication can still occur in lymphoid tissues. In addition, recent studies have suggested that adipose tissue provides an additional reservoir for HIV-infected macrophages and T lymphocytes even in the context of successful cART therapy. In this review article, we discuss possible mechanisms leading to the development of lymphoma in the cART era.
Collapse
|
156
|
Deeks SG, Lewin SR, Ross AL, Ananworanich J, Benkirane M, Cannon P, Chomont N, Douek D, Lifson JD, Lo YR, Kuritzkes D, Margolis D, Mellors J, Persaud D, Tucker JD, Barre-Sinoussi F, Alter G, Auerbach J, Autran B, Barouch DH, Behrens G, Cavazzana M, Chen Z, Cohen ÉA, Corbelli GM, Eholié S, Eyal N, Fidler S, Garcia L, Grossman C, Henderson G, Henrich TJ, Jefferys R, Kiem HP, McCune J, Moodley K, Newman PA, Nijhuis M, Nsubuga MS, Ott M, Palmer S, Richman D, Saez-Cirion A, Sharp M, Siliciano J, Silvestri G, Singh J, Spire B, Taylor J, Tolstrup M, Valente S, van Lunzen J, Walensky R, Wilson I, Zack J. International AIDS Society global scientific strategy: towards an HIV cure 2016. Nat Med 2016; 22:839-50. [PMID: 27400264 PMCID: PMC5322797 DOI: 10.1038/nm.4108] [Citation(s) in RCA: 369] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023]
Abstract
Antiretroviral therapy is not curative. Given the challenges in providing lifelong therapy to a global population of more than 35 million people living with HIV, there is intense interest in developing a cure for HIV infection. The International AIDS Society convened a group of international experts to develop a scientific strategy for research towards an HIV cure. This Perspective summarizes the group's strategy.
Collapse
Affiliation(s)
- Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Anna Laura Ross
- International and Scientific Relations Office, ANRS, Paris, France
| | - Jintanat Ananworanich
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Monsef Benkirane
- Molecular Virology Lab, Institute of Human Genetics, CNRS UPR 1142, Université de Montpellier, Montpellier, France
| | - Paula Cannon
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nicolas Chomont
- CRCHUM and Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, Montréal, Quebec, Canada
| | - Daniel Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Ying-Ru Lo
- World Health Organization Regional Office for the Western Pacific, Manila, Philippines
| | | | - David Margolis
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - John Mellors
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Persaud
- Johns Hopkins University School of Medicine &Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Joseph D Tucker
- University of North Carolina-Project China, Guangzhou, China
| | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Judith Auerbach
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Brigitte Autran
- Sorbonne Universités, UPMC Univ Paris 06, CIMI-Paris, France
- Inserm U1135, CIMI-Paris, Paris, France
- AP-HP, Hôpital Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Dan H Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Georg Behrens
- Clinic for Immunology and Rhematology, Hannover Medical School, Hanover, Germany
| | - Marina Cavazzana
- Centre d'Investigation Clinique en biothérapie, Hôpital Necker-Enfants Malades, Paris, France
| | - Zhiwei Chen
- AIDS Institute, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Éric A Cohen
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Quebec, Canada
| | | | - Serge Eholié
- Programme PAC-CI, Centre Hospitalier Universitaire de Treichville, Abidjan, Côte d'Ivoire
| | - Nir Eyal
- Harvard T. H. Chan School of Public Health, Department of Global Health and Population, Boston, Massachusetts, USA
| | - Sarah Fidler
- Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Cynthia Grossman
- National Institute of Mental Health, NIH, Bethesda, Maryland, USA
| | - Gail Henderson
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Timothy J Henrich
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Brigham &Women's Hospital, Boston, Massachusetts, USA
| | | | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joseph McCune
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Keymanthri Moodley
- Centre for Medical Ethics and Law, Department of Medicine, Stellenbosch University, Western Cape, South Africa
| | - Peter A Newman
- Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, Ontario, Canada
| | - Monique Nijhuis
- Department of Medical Microbiology, Virology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Melanie Ott
- Gladstone Institutes, University of California, San Francisco, San Francisco, California, USA
| | - Sarah Palmer
- Westmead Millennium Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Douglas Richman
- Virginia San Diego Healthcare System and University of California, San Diego, San Diego, California, USA
| | | | - Matthew Sharp
- Independent HIV Education and Advocacy Consultant, San Francisco, California, USA
| | - Janet Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guido Silvestri
- Yerkes National Primate Research Centre, Emory University, Atlanta, Georgia, USA
| | - Jerome Singh
- Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Jeffrey Taylor
- CARE Collaboratory Community Advisory Board, Palm Springs, California, USA
| | - Martin Tolstrup
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Susana Valente
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, Jupiter, Florida, USA
| | | | - Rochelle Walensky
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ira Wilson
- Department of Health Services, Policy &Practice, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Jerome Zack
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
157
|
Affiliation(s)
- Carola G. Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia;
| | - Michelle A. Linterman
- Lymphocyte Signalling and Development Institute Strategic Programme, Babraham Institute, Cambridge CB22 3AT, United Kingdom;
| | - Di Yu
- Laboratory for Molecular Immunomodulation, Department of Biochemistry and Molecular Biology, and Center for Inflammatory Diseases, Monash University, Melbourne, Victoria 3800, Australia;
| | - Ian C.M. MacLennan
- School of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
158
|
Miles B, Connick E. TFH in HIV Latency and as Sources of Replication-Competent Virus. Trends Microbiol 2016; 24:338-344. [PMID: 26947191 DOI: 10.1016/j.tim.2016.02.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 11/16/2022]
Abstract
During untreated disease, HIV replication is concentrated within T follicular helper cells (TFH). Heightened permissiveness, the presence of highly infectious virions on follicular dendritic cells (FDCs), low frequencies of virus-specific cytotoxic T lymphocytes (CTLs) in B cell follicles, expansions in TFH, and TFH dysfunction, all likely promote replication in TFH. Limited data suggest that memory TFH play a role in the latent or subclinical reservoir of HIV during antiretroviral therapy (ART), potentially for many of the same reasons. A better understanding of the role of memory TFH and FDC-bound virions in promoting recrudescent viremia in the setting of ART cessation is essential. Studies that target follicular virus reservoirs are needed to determine their role in HIV latency and to suggest successful cure strategies.
Collapse
Affiliation(s)
- Brodie Miles
- Division of Infectious Diseases, University of Colorado Denver, Aurora CO 80045, USA
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Colorado Denver, Aurora CO 80045, USA.
| |
Collapse
|
159
|
Massanella M, Fromentin R, Chomont N. Residual inflammation and viral reservoirs: alliance against an HIV cure. Curr Opin HIV AIDS 2016; 11:234-41. [PMID: 26575148 PMCID: PMC4743501 DOI: 10.1097/coh.0000000000000230] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW HIV persists in cellular and anatomical reservoirs during antiretroviral therapy (ART). Viral persistence is ensured by a variety of mechanisms including ongoing viral replication and proliferation of latently infected cells. In this review, we summarize recent findings establishing a link between the unresolved levels of inflammation observed in virally suppressed individuals on ART and the mechanisms responsible for HIV persistence. RECENT FINDINGS Residual levels of viral replication during ART are associated with persistent low levels of immune activation, suggesting that unresolved inflammation can promote the replenishment of the HIV reservoir in tissues. In addition, the recent findings that the latent HIV reservoir is maintained by continuous proliferation of latently infected cells provide another mechanism by which residual inflammation could contribute to HIV persistence. SUMMARY Residual inflammation during ART is likely to be a critical parameter contributing to HIV persistence. Therefore, reducing inflammation may be an efficient way to interfere with the maintenance of the HIV reservoir in virally suppressed individuals on ART.
Collapse
Affiliation(s)
- Marta Massanella
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| | - Rémi Fromentin
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| | - Nicolas Chomont
- Université de Montréal, Faculté de Médecine, Department of microbiology, infectiology and immunology, Montréal, QC, Canada
- Centre de Recherche du CHUM, Montréal, QC, Canada
| |
Collapse
|
160
|
Kohler SL, Pham MN, Folkvord JM, Arends T, Miller SM, Miles B, Meditz AL, McCarter M, Levy DN, Connick E. Germinal Center T Follicular Helper Cells Are Highly Permissive to HIV-1 and Alter Their Phenotype during Virus Replication. THE JOURNAL OF IMMUNOLOGY 2016; 196:2711-22. [PMID: 26873986 DOI: 10.4049/jimmunol.1502174] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/18/2016] [Indexed: 01/13/2023]
Abstract
HIV-1 replication is concentrated within CD4(+) T cells in B cell follicles of secondary lymphoid tissues during asymptomatic disease. Limited data suggest that a subset of T follicular helper cells (TFH) within germinal centers (GC) is highly permissive to HIV-1. Whether GC TFH are the major HIV-1 virus-producing cells in vivo has not been established. In this study, we investigated TFH permissivity to HIV-1 ex vivo by spinoculating and culturing tonsil cells with HIV-1 GFP reporter viruses. Using flow cytometry, higher percentages of GC TFH (CXCR5(high)PD-1(high)) and CXCR5(+)programmed cell death-1 (PD-1)(low) cells were GFP(+) than non-GC TFH (CXCR5(+)PD-1(intermediate)) or extrafollicular (EF) (CXCR5(-)) cells. When sorted prior to spinoculation, however, GC TFH were substantially more permissive than CXCR5(+)PD-1(low) or EF cells, suggesting that many GC TFH transition to a CXCR5(+)PD-1(low) phenotype during productive infection. In situ hybridization on inguinal lymph node sections from untreated HIV-1-infected individuals without AIDS revealed higher frequencies of HIV-1 RNA(+) cells in GC than non-GC regions of follicle or EF regions. Superinfection of HIV-1-infected individuals' lymph node cells with GFP reporter virus confirmed the permissivity of follicular cells ex vivo. Lymph node immunostaining revealed 96% of CXCR5(+)CD4(+) cells were located in follicles. Within sorted lymph node cells from four HIV-infected individuals, CXCR5(+) subsets harbored 11-66-fold more HIV-1 RNA than CXCR5(-) subsets, as determined by RT PCR. Thus, GC TFH are highly permissive to HIV-1, but downregulate PD-1 and, to a lesser extent, CXCR5 during HIV-1 replication. These data further implicate GC TFH as the major HIV-1-producing cells in chronic asymptomatic HIV-1 infection.
Collapse
Affiliation(s)
- Stephanie L Kohler
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael N Pham
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Joy M Folkvord
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Tessa Arends
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Shannon M Miller
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Brodie Miles
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Amie L Meditz
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Martin McCarter
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and
| | - David N Levy
- College of Dentistry, New York University, New York, NY 10010
| | - Elizabeth Connick
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045;
| |
Collapse
|
161
|
Vargas-Inchaustegui DA, Demers A, Shaw JM, Kang G, Ball D, Tuero I, Musich T, Mohanram V, Demberg T, Karpova TS, Li Q, Robert-Guroff M. Vaccine Induction of Lymph Node-Resident Simian Immunodeficiency Virus Env-Specific T Follicular Helper Cells in Rhesus Macaques. THE JOURNAL OF IMMUNOLOGY 2016; 196:1700-10. [PMID: 26773147 DOI: 10.4049/jimmunol.1502137] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/10/2015] [Indexed: 12/24/2022]
Abstract
Measurement of Ag-specific T follicular helper (TFH) cell activity in rhesus macaques has not previously been reported. Given that rhesus macaques are the animal model of choice for evaluating protective efficacy of HIV/SIV vaccine candidates and that TFH cells play a pivotal role in aiding B cell maturation, quantifying vaccine induction of HIV/SIV-specific TFH cells would greatly benefit vaccine development. In this study, we quantified SIV Env-specific IL-21-producing TFH cells for the first time, to our knowledge, in a nonhuman primate vaccine study. Macaques were primed twice mucosally with adenovirus 5 host range mutant recombinants encoding SIV Env, Rev, Gag, and Nef followed by two i.m. boosts with monomeric SIV gp120 or oligomeric SIV gp140 proteins. At 2 wk after the second protein boost, we obtained lymph node biopsy specimens and quantified the frequency of total and SIV Env-specific IL-21(+) TFH cells and total germinal center B cells, the size and number of germinal centers, and the frequency of SIV-specific Ab-secreting cells in B cell zones. Multiple correlation analyses established the importance of TFH for development of B cell responses in systemic and mucosally localized compartments, including blood, bone marrow, and rectum. Our results suggest that the SIV-specific TFH cells, initially induced by replicating adenovirus-recombinant priming, are long lived. The multiple correlations of SIV Env-specific TFH cells with systemic and mucosal SIV-specific B cell responses indicate that this cell population should be further investigated in HIV vaccine development as a novel correlate of immunity.
Collapse
Affiliation(s)
- Diego A Vargas-Inchaustegui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| | - Andrew Demers
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and
| | - Julia M Shaw
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Guobin Kang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and
| | - David Ball
- Center for Cancer Research Core Fluorescence Imaging Facility, Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Iskra Tuero
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Thomas Musich
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Venkatramanan Mohanram
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Thorsten Demberg
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tatiana S Karpova
- Center for Cancer Research Core Fluorescence Imaging Facility, Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583; and
| | - Marjorie Robert-Guroff
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
162
|
Towards an HIV cure based on targeted killing of infected cells: different approaches against acute versus chronic infection. Curr Opin HIV AIDS 2016; 10:207-13. [PMID: 25710815 DOI: 10.1097/coh.0000000000000151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Current regimens of combination antiretroviral therapy (cART) offer effective control of HIV infection, with maintenance of immune health and near-normal life expectancy. What will it take to progress beyond the status quo, whereby infectious virus can be eradicated (a 'sterilizing cure') or fully controlled without the need for ongoing cART (a 'functional cure')? RECENT FINDINGS On the basis of therapeutic advances in the cancer field, we propose that targeted cytotoxic therapy to kill HIV-infected cells represents a logical complement to cART for achieving an HIV cure. This concept is based on the fact that cART effectively blocks replication of the virus, but does not eliminate cells that are already infected; targeted cytotoxic therapy would contribute precisely this missing component. We suggest that different modalities are suited for curing primary acute versus established chronic infection. For acute infection, relatively short-acting potent agents such as recombinant immunotoxins might prove sufficient for HIV eradication, whereas for chronic infection, a long-lasting (lifelong?) modality is required to maintain full virus control, as might be achieved with genetically modified autologous T cells. SUMMARY We present perspectives for complementing cART with targeted cytotoxic therapy, whereby HIV infection is either eradicated or fully controlled, thereby eliminating the need for lifelong cART.
Collapse
|
163
|
Eckstrand CD, Hillman C, Smith AL, Sparger EE, Murphy BG. Viral Reservoirs in Lymph Nodes of FIV-Infected Progressor and Long-Term Non-Progressor Cats during the Asymptomatic Phase. PLoS One 2016; 11:e0146285. [PMID: 26741651 PMCID: PMC4704817 DOI: 10.1371/journal.pone.0146285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022] Open
Abstract
Background Examination of a cohort of cats experimentally infected with feline immunodeficiency virus (FIV) for 5.75 years revealed detectable proviral DNA in peripheral blood mononuclear cells (PBMCs) harvested during the asymptomatic phase, undetectable plasma viral RNA (FIV gag), and rarely detectable cell-associated viral RNA. Despite apparent viral latency in peripheral CD4+ T cells, circulating CD4+ T cell numbers progressively declined in progressor animals. The aim of this study was to explore this dichotomy of peripheral blood viral latency in the face of progressive immunopathology. The viral replication status, cellular immunophenotypes, and histopathologic features were compared between popliteal lymph nodes (PLNs) and peripheral blood. Also, we identified and further characterized one of the FIV-infected cats identified as a long-term non-progressor (LTNP). Results PLN-derived leukocytes from FIV-infected cats during the chronic asymptomatic phase demonstrated active viral gag transcription and FIV protein translation as determined by real-time RT-PCR, Western blot and in situ immunohistochemistry, whereas viral RNA in blood leukocytes was either undetectable or intermittently detectable and viral protein was not detected. Active transcription of viral RNA was detectable in PLN-derived CD4+ and CD21+ leukocytes. Replication competent provirus was reactivated ex vivo from PLN-derived leukocytes from three of four FIV-infected cats. Progressor cats showed a persistent and dramatically decreased proportion and absolute count of CD4+ T cells in blood, and a decreased proportion of CD4+ T cells in PLNs. A single long-term non-progressor (LTNP) cat persistently demonstrated an absolute peripheral blood CD4+ T cell count indistinguishable from uninfected animals, a lower proviral load in unfractionated blood and PLN leukocytes, and very low amounts of viral RNA in the PLN. Conclusion Collectively our data indicates that PLNs harbor important reservoirs of ongoing viral replication during the asymptomatic phase of infection, in spite of undetectable viral activity in peripheral blood. A thorough understanding of tissue-based lentiviral reservoirs is fundamental to medical interventions to eliminate virus or prolong the asymptomatic phase of FIV infection.
Collapse
Affiliation(s)
- C D Eckstrand
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - C Hillman
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - A L Smith
- Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - E E Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - B G Murphy
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| |
Collapse
|
164
|
Chan CN, Trinité B, Lee CS, Mahajan S, Anand A, Wodarz D, Sabbaj S, Bansal A, Goepfert PA, Levy DN. HIV-1 latency and virus production from unintegrated genomes following direct infection of resting CD4 T cells. Retrovirology 2016; 13:1. [PMID: 26728316 PMCID: PMC4700562 DOI: 10.1186/s12977-015-0234-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/21/2015] [Indexed: 12/31/2022] Open
Abstract
Background HIV-1 integration is prone to a high rate of failure, resulting in the accumulation of unintegrated viral genomes (uDNA) in vivo and in vitro. uDNA can be transcriptionally active, and circularized uDNA genomes are biochemically stable in non-proliferating cells. Resting, non-proliferating CD4 T cells are prime targets of HIV-1 infection and latently infected resting CD4 T cells are the major barrier to HIV cure. Our prior studies demonstrated that uDNA generates infectious virions when T cell activation follows rather than precedes infection. Results Here, we characterize in primary resting CD4 T cells the dynamics of integrated and unintegrated virus expression, genome persistence and sensitivity to latency reversing agents. Unintegrated HIV-1 was abundant in directly infected resting CD4 T cells. Maximal gene expression from uDNA was delayed compared with integrated HIV-1 and was less toxic, resulting in uDNA enrichment over time relative to integrated proviruses. Inhibiting integration with raltegravir shunted the generation of durable latency from integrated to unintegrated genomes. Latent uDNA was activated to de novo virus production by latency reversing agents that also activated latent integrated proviruses, including PKC activators, histone deacetylase inhibitors and P-TEFb agonists. However, uDNA responses displayed a wider dynamic range, indicating differential regulation of expression relative to integrated proviruses. Similar to what has recently been demonstrated for latent integrated proviruses, one or two applications of latency reversing agents failed to activate all latent unintegrated genomes. Unlike integrated proviruses, uDNA gene expression did not down modulate expression of HLA Class I on resting CD4 T cells. uDNA did, however, efficiently prime infected cells for killing by HIV-1-specific cytotoxic T cells. Conclusions These studies demonstrate that contributions by unintegrated genomes to HIV-1 gene expression, virus production, latency and immune responses are inherent properties of the direct infection of resting CD4 T cells. Experimental models of HIV-1 latency employing directly infected resting CD4 T cells should calibrate the contribution of unintegrated HIV-1. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0234-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chi N Chan
- Department of Basic Science, New York University College of Dentistry, New York, NY, 10010, USA.
| | - Benjamin Trinité
- Department of Basic Science, New York University College of Dentistry, New York, NY, 10010, USA.
| | - Caroline S Lee
- Department of Basic Science, New York University College of Dentistry, New York, NY, 10010, USA.
| | - Saurabh Mahajan
- Department of Basic Science, New York University College of Dentistry, New York, NY, 10010, USA.
| | - Akanksha Anand
- Department of Basic Science, New York University College of Dentistry, New York, NY, 10010, USA.
| | - Dominik Wodarz
- Department of Ecology and Evolutionary Biology, University of California, Irvine, School of Biological, Sciences, Irvine, CA, 92697, USA.
| | - Steffanie Sabbaj
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Anju Bansal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - David N Levy
- Department of Basic Science, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
165
|
Abstract
After the success of combination antiretroviral therapy (cART) to treat HIV infection, the next great frontier is to cure infected persons, a formidable challenge. HIV persists in a quiescent state in resting CD4+ T cells, where the replicative enzymes targeted by cART are not active. Although low levels of HIV transcripts are detectable in these resting cells, little to no viral protein is produced, rendering this reservoir difficult to detect by the host CD8+ T cell response. However, recent advances suggest that this state of latency might be pharmacologically reversed, resulting in viral protein expression without the adverse effects of massive cellular activation. Emerging data suggest that with this approach, infected cells will not die of viral cytopathic effects, but might be eliminated if HIV-specific CD8+ T cells can be effectively harnessed. Here, we address the antiviral properties of HIV-specific CD8+ T cells and how these cells might be harnessed to greater effect toward achieving viral eradication or a functional cure.
Collapse
|
166
|
Deleage C, Wietgrefe SW, Del Prete G, Morcock DR, Hao XP, Piatak M, Bess J, Anderson JL, Perkey KE, Reilly C, McCune JM, Haase AT, Lifson JD, Schacker TW, Estes JD. Defining HIV and SIV Reservoirs in Lymphoid Tissues. Pathog Immun 2016; 1:68-106. [PMID: 27430032 PMCID: PMC4943335 DOI: 10.20411/pai.v1i1.100] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A primary obstacle to an HIV-1 cure is long-lived viral reservoirs, which must be eliminated or greatly reduced. Cure strategies have largely focused on monitoring changes in T cell reservoirs in peripheral blood (PB), even though the lymphoid tissues (LT) are primary sites for viral persistence. To track and discriminate viral reservoirs within tissue compartments we developed a specific and sensitive next-generation in situ hybridization approach to detect vRNA, including vRNA+ cells and viral particles ("RNAscope"), vDNA+ cells ("DNAscope") and combined vRNA and vDNA with immunohistochemistry to detect and phenotype active and latently infected cells in the same tissue section. RNAscope is highly sensitive with greater speed of analysis compared to traditional in situ hybridization. The highly sensitive and specific DNAscope detected SIV/HIV vDNA+ cells, including duplexed detection of vDNA and vRNA or immunophenotypic markers in the same section. Analysis of LT samples from macaques prior to and during combination antiretroviral therapy demonstrated that B cell follicles are an important anatomical compartment for both latent and active viral persistence during treatment. These new tools should allow new insights into viral reservoir biology and evaluation of cure strategies.
Collapse
Affiliation(s)
- Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Stephen W. Wietgrefe
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Gregory Del Prete
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - David R. Morcock
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Xing Pei Hao
- Pathology and Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
- Deceased 19 September 2014
| | - Julian Bess
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jodi L. Anderson
- Department of Medicine. Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Katherine E. Perkey
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Cavan Reilly
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| | - Ashley T. Haase
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Timothy W. Schacker
- Department of Medicine. Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
167
|
Heesters BA, Lindqvist M, Vagefi PA, Scully EP, Schildberg FA, Altfeld M, Walker BD, Kaufmann DE, Carroll MC. Follicular Dendritic Cells Retain Infectious HIV in Cycling Endosomes. PLoS Pathog 2015; 11:e1005285. [PMID: 26623655 PMCID: PMC4666623 DOI: 10.1371/journal.ppat.1005285] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/27/2015] [Indexed: 01/12/2023] Open
Abstract
Despite the success of antiretroviral therapy (ART), it does not cure Human Immunodeficiency Virus (HIV) and discontinuation results in viral rebound. Follicular dendritic cells (FDC) are in direct contact with CD4+ T cells and they retain intact antigen for prolonged periods. We found that human FDC isolated from patients on ART retain infectious HIV within a non-degradative cycling compartment and transmit infectious virus to uninfected CD4 T cells in vitro. Importantly, treatment of the HIV+ FDC with a soluble complement receptor 2 purges the FDC of HIV virions and prevents viral transmission in vitro. Our results provide an explanation for how FDC can retain infectious HIV for extended periods and suggest a therapeutic strategy to achieve cure in HIV-infected humans. Human immunodeficiency virus (HIV) can lead to acquired immunodeficiency syndrome, or AIDS. Before the introduction of anti retroviral therapy (ART) in the mid-1990s, people with HIV could progress to AIDS in just a few years. Today patients with HIV have a close to normal life expectancy. Worldwide, there are about 2 million new cases of HIV per year. Currently about 35 million people are living with HIV of which around 13 million receive ART. Still an estimated 1.5 million people die from the consequences of HIV each year. Despite the success of ART, it does not cure HIV and discontinuation results in viral rebound. Follicular dendritic cells (FDC), located central to the B cell follicle, are also in direct contact with T cells. FDCs retain intact antigen for prolonged periods. We found that human FDCs isolated from patients on ART retain infectious HIV and can transmit virus to uninfected T cells in vitro. Treatment of the HIV+ FDC with a soluble complement receptor 2 purges the FDC of HIV virions and prevents viral transmission to T cells in vitro. Our results can explain how FDCs retain infectious HIV and suggest a therapeutic strategy to come closer to a cure.
Collapse
Affiliation(s)
- Balthasar A. Heesters
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands
- * E-mail:
| | - Madelene Lindqvist
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Center and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
| | - Parsia A. Vagefi
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Eileen P. Scully
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Frank A. Schildberg
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marcus Altfeld
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Department of Viral Immunology, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Bruce D. Walker
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Center and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
| | - Daniel E. Kaufmann
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Center and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
- Centre de Recherché du CHUM; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Michael C. Carroll
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
168
|
Persistent Simian Immunodeficiency Virus Infection Drives Differentiation, Aberrant Accumulation, and Latent Infection of Germinal Center Follicular T Helper Cells. J Virol 2015; 90:1578-87. [PMID: 26608323 DOI: 10.1128/jvi.02471-15] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/17/2015] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED CD4(+) follicular T helper (Tfh) cells play a prominent role in humoral immune responses, but the mechanisms of their accumulation and infection in AIDS remain unclear. Here we found that germinal center (GC) Tfh cells, defined here as CXCR5(+) PD-1(HIGH) CD4(+) T cells, do not express the HIV coreceptor CCR5 yet serve as a latent reservoir in GCs. With disease progression, an expansion of GC Tfh cells is accompanied by increases in dysfunctional CD8(+) T cells. In contrast, Tfh precursor (CXCR5(-) CD4(+) T) cells in lymph nodes do express CCR5 and differentiate into GC Tfh cells following interleukin-6 (IL-6) and IL-21 stimulation, and viral DNA is detectable in fully differentiated GC Tfh cells ex vivo. This suggests that SIV-infected GC Tfh cells may be derived from Tfh precursor cell subsets that become infected in marginal zones and then migrate into GCs as fully mature GC Tfh cells that serve as persistent virus reservoirs. These findings suggest that viral persistence in lymph nodes drives compensatory differentiation, aberrant accumulation, and latent infection of GC Tfh cells, resulting in marked impairment of humoral immune responses. IMPORTANCE Generation of antibodies that can effectively eliminate viruses requires interactions of B cells with highly specialized T cells in GCs of lymphoid tissues called follicular T helper cells. Here we show that in simian immunodeficiency virus infection, these cells are initially infected in a precursor stage that leads to alterations in their homing, accumulation, and function that may be responsible for the inability of human immunodeficiency virus-infected patients to generate effective antibody responses.
Collapse
|
169
|
Brockman MA, Jones RB, Brumme ZL. Challenges and Opportunities for T-Cell-Mediated Strategies to Eliminate HIV Reservoirs. Front Immunol 2015; 6:506. [PMID: 26483795 PMCID: PMC4591506 DOI: 10.3389/fimmu.2015.00506] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/17/2015] [Indexed: 12/17/2022] Open
Abstract
HIV's ability to establish latent reservoirs of reactivation-competent virus is the major barrier to cure. "Shock and kill" methods consisting of latency-reversing agents (LRAs) followed by elimination of reactivating cells through cytopathic effects are under active development. However, the clinical efficacy of LRAs remains to be established. Moreover, recent studies indicate that reservoirs may not be reduced efficiently by either viral cytopathic or CD8(+) T-cell-mediated mechanisms. In this perspective, we highlight challenges to T-cell-mediated elimination of HIV reservoirs, including characteristics of responding T cells, aspects of the cellular reservoirs, and properties of the latent virus itself. We also discuss potential strategies to overcome these challenges by targeting the antiviral activity of T cells toward appropriate viral antigens following latency.
Collapse
Affiliation(s)
- Mark A Brockman
- Faculty of Health Sciences, Simon Fraser University , Burnaby, BC , Canada ; BC Centre for Excellence in HIV/AIDS , Vancouver, BC , Canada
| | - R Brad Jones
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University , Washington, DC , USA
| | - Zabrina L Brumme
- Faculty of Health Sciences, Simon Fraser University , Burnaby, BC , Canada ; BC Centre for Excellence in HIV/AIDS , Vancouver, BC , Canada
| |
Collapse
|
170
|
Affiliation(s)
- Hendrik Streeck
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA, and at the Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| |
Collapse
|
171
|
Abstract
PURPOSE OF REVIEW T follicular helper (Tfh) cells play a critical role as providers of B-cell help and dysfunction in Tfh/B-cell interactions can lead to autoimmunity or immunodeficiency. These observations have generated a great deal of interest in understanding how these cells are affected during HIV infection and how their functional changes might affect antibody responses. RECENT FINDINGS Recent studies have shown that HIV/simian immunodeficiency virus (SIV) infection affects both Tfh-cell frequency and function and suggest that Tfh-cell perturbations might contribute to the relative inefficiency of HIV-infected individuals to generate broadly neutralizing antibodies (bNAbs). SUMMARY The present review will highlight these recent findings addressing the role of Tfh cells in HIV infection as well as the impact HIV infection has on Tfh and circulating memory Tfh (cTfh) cell frequency and function.
Collapse
|
172
|
Haas MK, Levy DN, Folkvord JM, Connick E. Distinct patterns of Bcl-2 expression occur in R5- and X4-tropic HIV-1-producing lymphoid tissue cells infected ex vivo. AIDS Res Hum Retroviruses 2015; 31:298-304. [PMID: 25353356 DOI: 10.1089/aid.2014.0155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Most HIV-1 replication occurs in secondary lymphoid tissues in T cells within B cell follicles. Mechanisms underlying the accumulation of HIV-1-producing cells at these sites are not understood. Antiapoptotic proteins such as Bcl-2 could promote follicular CD4(+) T cell survival, contributing to sustained virus production. Tonsils obtained from subjects without known HIV infection were disaggregated and analyzed for Bcl-2 expression in follicular (CXCR5(+)) and extrafollicular (CXCR5(-)) CD3(+)CD4(+) cells by flow cytometry. Additional tonsil cells were cultured with phytohemagglutinin (PHA) and interleukin-2 (IL-2) for 2 days, infected with either CCR5(R5) or CXCR4-tropic (X4) GFP reporter viruses, and analyzed for Bcl-2 expression. In freshly disaggregated CD3(+)CD4(+) tonsil cells, mean florescence intensity (MFI) for Bcl-2 was higher in CXCR5(+) (median, 292) compared to CXCR5(-) cells (median, 194; p=0.001). Following in vitro stimulation with PHA and IL-2, Bcl-2 MFI was higher in both CXCR5(+) cells (median, 757; p=0.03) and CXCR5(-) cells (median, 884; p=0.002) in uninfected cultures compared to freshly isolated tonsil cells. Bcl-2 MFI was higher in GFP(+)CD3(+)CD8(-) R5-producing cells (median, 554) than in X4-producing cells (median, 393; p=0.02). Bcl-2 MFI was higher in R5-producing CXCR5(+) cells (median, 840) compared to all other subsets including R5-producing CXCR5(-) cells (median, 524; p=0.04), X4-producing CXCR5(+) cells (median, 401; p=0.02), and X4-producing CXCR5(-) cells (median, 332; p=0.008). Bcl-2 expression is elevated in R5 HIV-1-producing CXCR5(+) T cells in vitro, which may contribute to propagation of R5 virus in B cell follicles in vivo.
Collapse
Affiliation(s)
- Michelle K. Haas
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
- Denver Public Health, Denver, Colorado
| | - David N. Levy
- College of Dentistry, New York University, New York, New York
| | - Joy M. Folkvord
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Elizabeth Connick
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| |
Collapse
|
173
|
Fukazawa Y, Lum R, Okoye AA, Park H, Matsuda K, Bae JY, Hagen SI, Shoemaker R, Deleage C, Lucero C, Morcock D, Swanson T, Legasse AW, Axthelm MK, Hesselgesser J, Geleziunas R, Hirsch VM, Edlefsen PT, Piatak M, Estes JD, Lifson JD, Picker LJ. B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in elite controllers. Nat Med 2015; 21:132-9. [PMID: 25599132 PMCID: PMC4320022 DOI: 10.1038/nm.3781] [Citation(s) in RCA: 410] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/30/2014] [Indexed: 12/17/2022]
Abstract
Chronic-phase HIV and simian immunodeficiency virus (SIV) replication is reduced by as much as 10,000-fold in elite controllers (ECs) compared with typical progressors (TPs), but sufficient viral replication persists in EC tissues to allow viral sequence evolution and induce excess immune activation. Here we show that productive SIV infection in rhesus monkey ECs, but not TPs, is markedly restricted to CD4(+) follicular helper T (TFH) cells, suggesting that these EC monkeys' highly effective SIV-specific CD8(+) T cells can effectively clear productive SIV infection from extrafollicular sites, but their relative exclusion from B cell follicles prevents their elimination of productively infected TFH cells. CD8(+) lymphocyte depletion in EC monkeys resulted in a dramatic re-distribution of productive SIV infection to non-TFH cells, with restriction of productive infection to TFH cells resuming upon CD8(+) T cell recovery. Thus, B cell follicles constitute 'sanctuaries' for persistent SIV replication in the presence of potent anti-viral CD8(+) T cell responses, potentially complicating efforts to cure HIV infection with therapeutic vaccination or T cell immunotherapy.
Collapse
Affiliation(s)
- Yoshinori Fukazawa
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Richard Lum
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Afam A. Okoye
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Haesun Park
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Kenta Matsuda
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jin Young Bae
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Shoko I. Hagen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Carissa Lucero
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - David Morcock
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Tonya Swanson
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Alfred W. Legasse
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | | | | | - Vanessa M. Hirsch
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Paul T. Edlefsen
- Statistical Center for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| |
Collapse
|
174
|
Abdelaal HM, Kim HO, Wagstaff R, Sawahata R, Southern PJ, Skinner PJ. Comparison of Vibratome and Compresstome sectioning of fresh primate lymphoid and genital tissues for in situ MHC-tetramer and immunofluorescence staining. Biol Proced Online 2015; 17:2. [PMID: 25657614 PMCID: PMC4318225 DOI: 10.1186/s12575-014-0012-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/29/2014] [Indexed: 11/10/2022] Open
Abstract
Background For decades, the Vibratome served as a standard laboratory resource for sectioning fresh and fixed tissues. In skilled hands, high quality and consistent fresh unfixed tissue sections can be produced using a Vibratome but the sectioning procedure is extremely time consuming. In this study, we conducted a systematic comparison between the Vibratome and a new approach to section fresh unfixed tissues using a Compresstome. We used a Vibratome and a Compresstome to cut fresh unfixed lymphoid and genital non-human primate tissues then used in situ tetramer staining to label virus-specific CD8 T cells and immunofluorescent counter-staining to label B and T cells. We compared the Vibratome and Compresstome in five different sectioning parameters: speed of cutting, chilling capability, specimen stabilization, size of section, and section/staining quality. Results Overall, the Compresstome and Vibratome both produced high quality sections from unfixed spleen, lymph node, vagina, cervix, and uterus, and subsequent immunofluorescent staining was equivalent. The Compresstome however, offered distinct advantages; producing sections approximately 5 times faster than the Vibratome, cutting tissue sections more easily, and allowing production of larger sections. Conclusions A Compresstome can be used to generate fresh unfixed primate lymph node, spleen, vagina, cervix and uterus sections, and is superior to a Vibratome in cutting these fresh tissues.
Collapse
Affiliation(s)
- Hadia M Abdelaal
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA ; Departments of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519 Egypt
| | - Hyeon O Kim
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| | - Reece Wagstaff
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| | - Ryoko Sawahata
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| | - Peter J Southern
- Departments of Microbiology, MMC 196, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455 USA
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| |
Collapse
|
175
|
Connick E, Folkvord JM, Lind KT, Rakasz EG, Miles B, Wilson NA, Santiago ML, Schmitt K, Stephens EB, Kim HO, Wagstaff R, Li S, Abdelaal HM, Kemp N, Watkins DI, MaWhinney S, Skinner PJ. Compartmentalization of simian immunodeficiency virus replication within secondary lymphoid tissues of rhesus macaques is linked to disease stage and inversely related to localization of virus-specific CTL. THE JOURNAL OF IMMUNOLOGY 2014; 193:5613-25. [PMID: 25362178 DOI: 10.4049/jimmunol.1401161] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We previously demonstrated that HIV replication is concentrated in lymph node B cell follicles during chronic infection and that HIV-specific CTL fail to accumulate in large numbers at those sites. It is unknown whether these observations can be generalized to other secondary lymphoid tissues or whether virus compartmentalization occurs in the absence of CTL. We evaluated these questions in SIVmac239-infected rhesus macaques by quantifying SIV RNA(+) cells and SIV-specific CTL in situ in spleen, lymph nodes, and intestinal tissues obtained at several stages of infection. During chronic asymptomatic infection prior to simian AIDS, SIV-producing cells were more concentrated in follicular (F) compared with extrafollicular (EF) regions of secondary lymphoid tissues. At day 14 of infection, when CTL have minimal impact on virus replication, there was no compartmentalization of SIV-producing cells. Virus compartmentalization was diminished in animals with simian AIDS, which often have low-frequency CTL responses. SIV-specific CTL were consistently more concentrated within EF regions of lymph node and spleen in chronically infected animals regardless of epitope specificity. Frequencies of SIV-specific CTL within F and EF compartments predicted SIV RNA(+) cells within these compartments in a mixed model. Few SIV-specific CTL expressed the F homing molecule CXCR5 in the absence of the EF retention molecule CCR7, possibly accounting for the paucity of F CTL. These findings bolster the hypothesis that B cell follicles are immune privileged sites and suggest that strategies to augment CTL in B cell follicles could lead to improved viral control and possibly a functional cure for HIV infection.
Collapse
Affiliation(s)
- Elizabeth Connick
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045;
| | - Joy M Folkvord
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045
| | - Katherine T Lind
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| | - Brodie Miles
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045
| | - Nancy A Wilson
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| | - Mario L Santiago
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045
| | - Kimberly Schmitt
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Edward B Stephens
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Hyeon O Kim
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Reece Wagstaff
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Shengbin Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Hadia M Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN; Department of Microbiology and Immunology, Zagazig University, Zagazig, Egypt 44519; and
| | - Nathan Kemp
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - David I Watkins
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715
| | - Samantha MaWhinney
- Department of Biostatistics and Informatics, University of Colorado Denver, Aurora, CO 80045
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| |
Collapse
|
176
|
Kevin Ward CJ. Defining and targeting residual virus on combined antiretroviral therapy. Future Virol 2014. [DOI: 10.2217/fvl.14.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
20th International AIDS Conference Melbourne, Australia, 20–25 July 2014
Collapse
|
177
|
Hong JJ, Amancha PK, Rogers KA, Courtney CL, Havenar-Daughton C, Crotty S, Ansari AA, Villinger F. Early lymphoid responses and germinal center formation correlate with lower viral load set points and better prognosis of simian immunodeficiency virus infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:797-806. [PMID: 24907346 DOI: 10.4049/jimmunol.1400749] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We have investigated the dynamics of germinal center (GC) formation in lymphoid tissues following acute SIV infection. SIV induces a marked follicular hyperplasia, associated with an aberrant accumulation of nonproliferating T follicular helper cells within GCs, but with an abundance of cells producing IL-21, demonstrating that the mechanisms involved for these two events appear independent. IL-21-stimulated T follicular helper cells are considered a critical element for GC formation, a physiological process that seems dysregulated and excessive during HIV/SIV infection, contributing to lymphoid pathogenesis. However, the data suggest that the kinetics by which such GCs are formed may be an important predictor of the host-pathogen equilibrium, as early GC hyperplasia was associated with better control of viral replication. In contrast, monkeys undergoing fast disease progression upon infection exhibited an involution of GCs without local IL-21 production in GCs. These results provide important clues regarding GC-related hyperimmune responses in the context of disease progression within various individuals during HIV/SIV infection and may open novel therapeutic avenues to limit lymphoid dysfunction, postinfection.
Collapse
Affiliation(s)
- Jung Joo Hong
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322; Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Praveen K Amancha
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322; Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Kenneth A Rogers
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322; Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Cynthia L Courtney
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322; Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Colin Havenar-Daughton
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Aftab A Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322; Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Francois Villinger
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322; Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| |
Collapse
|
178
|
Meditz AL, Connick E, McCarter M. Safety of excisional inguinal lymph node biopsies performed for research purposes in HIV-1-infected women and men. Surg Infect (Larchmt) 2014; 15:399-403. [PMID: 24815090 DOI: 10.1089/sur.2013.218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Most HIV-1 replication occurs in secondary lymphoid tissues, and evaluating these tissues is crucial to investigations of pathogenesis. Inguinal lymph nodes (LN) are obtained frequently for these studies as they are readily detectable in most individuals and provide abundant numbers of cells. Knowledge of the outcomes of inguinal LN excision for research purposes is important to inform accurately study participants and researchers of the potential risks. METHODS Data on surgical complications were collected in real time in HIV-1-infected subjects who underwent excisional inguinal LN biopsies for research purposes from February 1997 through June 2011. Data were analyzed retrospectively to determine the frequency of surgical complications using the Fisher exact test and non-parametric testing. RESULTS Eighty-seven research subjects underwent a total of 95 LN excisions. Thirty-six percent of subjects were female, 53% were white, 26% were black, 16% Hispanic, and 2% Native American. Median age was 36 y (22-52). The median CD4+ T cell count was 478 cell/mm(3) (range, 57-1117) and the median plasma HIV-1 RNA concentration was 4.1 log10copies/mL (range, 1.7-5.9). Minor complications including seroma, transient lymphedema, hematoma, and allergic reaction to surgical tape, occurred in 10% of procedures. Complications that required medical attention occurred in an additional 10% of procedures, and included cellulitis (5%), superficial incisional surgical site infection (3%), and seroma requiring aspiration (1%). Subjects with complications had a lower BMI (25; range, 16-38; n=12) than others (28; range, 19-57; n=40; p=0.05) and tended to have higher platelets, (median, 259×10(9)/L; range, 196-332; vs. 233×10(9)/L; range, 44-633; p=0.07). No other clinical or laboratory characteristics were associated with complications (p≥0.3). CONCLUSIONS Lymph node excision for research purposes is generally safe in a diverse group of chronically HIV-1-infected women and men, but can result in complications in a minority of subjects. No predictors of complications were identified.
Collapse
Affiliation(s)
- Amie L Meditz
- 1 Division of Infectious Diseases, Department of Medicine, University of Colorado-Denver , Aurora, Colorado
| | | | | |
Collapse
|
179
|
Demers KR, Reuter MA, Betts MR. CD8(+) T-cell effector function and transcriptional regulation during HIV pathogenesis. Immunol Rev 2013; 254:190-206. [PMID: 23772621 PMCID: PMC3693771 DOI: 10.1111/imr.12069] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A detailed understanding of the immune response to human immunodeficiency virus (HIV) infection is needed to inform prevention and therapeutic strategies that aim to contain the acquired immunodeficiency syndrome (AIDS) pandemic. The cellular immune response plays a critical role in controlling viral replication during HIV infection and will likely need to be a part of any vaccine approach. The qualitative feature of the cellular response most closely associated with immunological control of HIV infection is CD8(+) T-cell cytotoxic potential, which is responsible for mediating the elimination of infected CD4(+) T cells. Understanding the underlying mechanisms involved in regulating the elicitation and maintenance of this kind of effector response can provide guidance for vaccine design. In this review, we discuss the evidence for CD8(+) T cells as correlates of protection, the means by which their antiviral capacity is evaluated, and transcription factors responsible for their function, or dysfunction, during HIV infection.
Collapse
Affiliation(s)
- Korey R. Demers
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Morgan A. Reuter
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael R. Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
180
|
Perreau M, Savoye AL, De Crignis E, Corpataux JM, Cubas R, Haddad EK, De Leval L, Graziosi C, Pantaleo G. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. ACTA ACUST UNITED AC 2012; 210:143-56. [PMID: 23254284 PMCID: PMC3549706 DOI: 10.1084/jem.20121932] [Citation(s) in RCA: 534] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Follicular T helper cells are the major reservoir for HIV infection and accumulate during chronic HIV infection. In the present study, we have investigated the distribution of HIV-specific and HIV-infected CD4 T cells within different populations of memory CD4 T cells isolated from lymph nodes of viremic HIV-infected subjects. Four memory CD4 T cell populations were identified on the basis of the expression of CXCR5, PD-1, and Bcl-6: CXCR5−PD-1−Bcl-6−, CXCR5+PD-1−Bcl-6−, CXCR5−PD-1+Bcl-6−, and CXCR5+PD-1+Bcl-6+. On the basis of Bcl-6 expression and functional properties (IL-21 production and B cell help), the CXCR5+PD-1+Bcl-6+ cell population was considered to correspond to the T follicular helper (Tfh) cell population. We show that Tfh and CXCR5−PD-1+ cell populations are enriched in HIV-specific CD4 T cells, and these populations are significantly increased in viremic HIV-infected subjects as compared with healthy subjects. The Tfh cell population contained the highest percentage of CD4 T cells harboring HIV DNA and was the most efficient in supporting productive infection in vitro. Replication competent HIV was also readily isolated from Tfh cells in subjects with nonprogressive infection and low viremia (<1,000 HIV RNA copies). However, only the percentage of Tfh cells correlated with the levels of plasma viremia. These results demonstrate that Tfh cells serve as the major CD4 T cell compartment for HIV infection, replication, and production.
Collapse
Affiliation(s)
- Matthieu Perreau
- Divisions of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Riou C, Ganusov VV, Campion S, Mlotshwa M, Liu MKP, Whale VE, Goonetilleke N, Borrow P, Ferrari G, Betts MR, Haynes BF, McMichael AJ, Gray CM. Distinct kinetics of Gag-specific CD4+ and CD8+ T cell responses during acute HIV-1 infection. THE JOURNAL OF IMMUNOLOGY 2012; 188:2198-206. [PMID: 22287716 DOI: 10.4049/jimmunol.1102813] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
HIV infection is characterized by a gradual deterioration of immune function, mainly in the CD4 compartment. To better understand the dynamics of HIV-specific T cells, we analyzed the kinetics and polyfunctional profiles of Gag-specific CD4(+) and CD8(+) T cell responses in 12 subtype C-infected individuals with different disease-progression profiles, ranging from acute to chronic HIV infection. The frequencies of Gag-responsive CD4(+) and CD8(+) T cells showed distinct temporal kinetics. The peak frequency of Gag-responsive IFN-γ(+)CD4(+) T cells was observed at a median of 28 d (interquartile range: 21-81 d) post-Fiebig I/II staging, whereas Gag-specific IFN-γ(+)CD8(+) T cell responses peaked at a median of 253 d (interquartile range: 136-401 d) and showed a significant biphasic expansion. The proportion of TNF-α-expressing cells within the IFN-γ(+)CD4(+) T cell population increased (p = 0.001) over time, whereas TNF-α-expressing cells within IFN-γ(+)CD8(+) T cells declined (p = 0.005). Both Gag-responsive CD4(+) and CD8(+) T cells showed decreased Ki67 expression within the first 120 d post-Fiebig I/II staging. Prior to the disappearance of Gag-responsive Ki67(+)CD4(+) T cells, these cells positively correlated (p = 0.00038) with viremia, indicating that early Gag-responsive CD4 events are shaped by viral burden. No such associations were observed in the Gag-specific CD8(+) T cell compartment. Overall, these observations indicated that circulating Gag-responsive CD4(+) and CD8(+) T cell frequencies and functions are not synchronous, and properties change rapidly at different tempos during early HIV infection.
Collapse
Affiliation(s)
- Catherine Riou
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town 7529, South Africa
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Horiike M, Iwami S, Kodama M, Sato A, Watanabe Y, Yasui M, Ishida Y, Kobayashi T, Miura T, Igarashi T. Lymph nodes harbor viral reservoirs that cause rebound of plasma viremia in SIV-infected macaques upon cessation of combined antiretroviral therapy. Virology 2011; 423:107-18. [PMID: 22196013 DOI: 10.1016/j.virol.2011.11.024] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/06/2011] [Accepted: 11/29/2011] [Indexed: 12/16/2022]
Abstract
Attempts to find a cure for HIV infection are hindered by the presence of viral reservoirs that resist highly active antiretroviral therapy. To identify the properties of these reservoirs, four SIV239-infected Rhesus macaques were treated with combined antiretroviral therapy (cART) for 1 year. While plasma viral RNA (vRNA) was effectively suppressed, a systemic analysis revealed that vRNA was distributed in the following order: lymphatic tissues>lungs and intestine>other tissues. Histochemistry yielded no cells with viral signals. To increase the chance of detection, two additional SIV-infected animals were treated and analyzed on Day 10 after the cessation of cART. These animals exhibited similar vRNA distribution patterns to the former animals, and immunohistochemistry revealed Nef-positive T lymphocytes predominantly in the follicles of mesenteric lymph nodes (MLNs). These data suggest that lymphatic tissues, including MLNs, contain major cellular reservoirs that cause rebound of plasma viremia upon cessation of therapy.
Collapse
Affiliation(s)
- Mariko Horiike
- Laboratory of Primate Model, Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Meythaler M, Wang Z, Martinot A, Pryputniewicz S, Kasheta M, McClure HM, O’Neil SP, Kaur A. Early induction of polyfunctional simian immunodeficiency virus (SIV)-specific T lymphocytes and rapid disappearance of SIV from lymph nodes of sooty mangabeys during primary infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:5151-61. [PMID: 21441446 PMCID: PMC3130630 DOI: 10.4049/jimmunol.1004110] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although the cellular immune response is essential for controlling SIV replication in Asian macaques, its role in maintaining nonpathogenic SIV infection in natural hosts such as sooty mangabeys (SM) remains to be defined. We have previously shown that similar to rhesus macaques (RM), SM are able to mount a T lymphocyte response against SIV infection. To investigate early control of SIV replication in natural hosts, we performed a detailed characterization of SIV-specific cellular immunity and viral control in the first 6 mo following SIV infection in SM. Detection of the initial SIV-specific IFN-γ ELISPOT response in SIVsmE041-infected SM coincided temporally with a decline in peak plasma viremia and was similar in magnitude, specificity, and breadth to SIVsmE041-infected and SIVmac239-infected RM. Despite these similarities, SM showed a greater reduction in postpeak plasma viremia and a more rapid disappearance of productively SIV-infected cells from the lymph node compared with SIVmac239-infected RM. The early Gag-specific CD8(+) T lymphocyte response was significantly more polyfunctional in SM compared with RM, and granzyme B-positive CD8(+) T lymphocytes were present at significantly higher frequencies in SM even prior to SIV infection. These findings suggest that the early SIV-specific T cell response may be an important determinant of lymphoid tissue viral clearance and absence of lymph node immunopathology in natural hosts of SIV infection.
Collapse
Affiliation(s)
- Mareike Meythaler
- Department of Immunology, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772
| | - Zichun Wang
- Department of Immunology, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772
| | - Amanda Martinot
- Comparative Pathology New England Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772
| | - Sarah Pryputniewicz
- Department of Immunology, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772
| | - Melissa Kasheta
- Department of Immunology, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772
| | - Harold M. McClure
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30322
| | - Shawn P. O’Neil
- Comparative Pathology New England Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772
| | - Amitinder Kaur
- Department of Immunology, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772
| |
Collapse
|
184
|
Reynolds MR, Piaskowski SM, Weisgrau KL, Weiler AM, Friedrich TC, Rakasz EG. Ex vivo analysis of SIV-infected cells by flow cytometry. Cytometry A 2011; 77:1059-66. [PMID: 20722008 DOI: 10.1002/cyto.a.20960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Deciphering the complex interactions between human and simian immunodeficiency viruses (HIV/SIV) and their host cells is crucial to the development of improved therapies and vaccines. Investigating these relationships has been complicated by the inability to directly analyze infected cells among freshly isolated peripheral blood lymphocytes. Here, we describe a method to detect cells productively infected with SIVmac239 ex vivo from the blood or lymph nodes by flow cytometry. Using this method, we show a close correlation between the frequency of productively infected cells in both sample type and the plasma viral load. We define that the minimum threshold for detecting productively infected cells in lymph nodes by flow cytometry requires a plasma virus concentration of ∼2.5 × 10(4) vRNA copy Equivalents (Eq)/ml. Conversely, an approximately 2 logs higher plasma viral load is needed to detect productively infected cells in the peripheral blood. This novel protocol provides a direct analytical tool to assess interactions between SIV and host cells, which is of key importance to investigators in AIDS research.
Collapse
Affiliation(s)
- Matthew R Reynolds
- AIDS Vaccine Research Laboratory, University of Wisconsin-Madison, Madison, Wisconsin 53711, USA
| | | | | | | | | | | |
Collapse
|
185
|
Schooley RT, Spritzler J, Wang H, Lederman MM, Havlir D, Kuritzkes DR, Pollard R, Battaglia C, Robertson M, Mehrotra D, Casimiro D, Cox K, Schock B, AIDS Clinical Trials Group 5197 Study Team. AIDS clinical trials group 5197: a placebo-controlled trial of immunization of HIV-1-infected persons with a replication-deficient adenovirus type 5 vaccine expressing the HIV-1 core protein. J Infect Dis 2010; 202:705-16. [PMID: 20662716 PMCID: PMC2916952 DOI: 10.1086/655468] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1)-specific cellular immunity contributes to the control of HIV-1 replication. HIV-1-infected volunteers who were receiving antiretroviral therapy were given a replication-defective adenovirus type 5 HIV-1 gag vaccine in a randomized, blinded therapeutic vaccination study. METHODS HIV-1-infected vaccine or placebo recipients underwent analytical treatment interruption (ATI) for 16 weeks. The log(10) HIV-1 RNA load at the ATI set point and the time-averaged area under the curve served as co-primary end points. Immune responses were measured by intracellular cytokine staining and carboxyfluorescein succinimidyl ester dye dilution. RESULTS Vaccine benefit trends were seen for both primary end points, but they did not reach a prespecified significance level of P < or = 25. The estimated shifts in the time-averaged area under the curve and the ATI set point were 0.24 (P=.04, unadjusted) and 0.26 (P=.07, unadjusted) log(10) copies lower, respectively, in the vaccine arm than in the placebo arm. HIV-1 gag-specific CD4(+) cells producing interferon-gamma were an immunologic correlate of viral control. CONCLUSION The vaccine was generally safe and well tolerated. Despite a trend favoring viral suppression among vaccine recipients, differences in HIV-1 RNA levels did not meet the prespecified level of significance. Induction of HIV-1 gag-specific CD4 cells correlated with control of viral replication in vivo. Future immunogenicity studies should require a substantially higher immunogenicity threshold before an ATI is contemplated.
Collapse
Affiliation(s)
- Robert T Schooley
- Div. of Infectious Diseases,University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
Collaborators
Linda Meixner, Susan Cahill, Trisha Walton, Barbara Gripshover, Sue Richard, Kelley Carpenter, David M Asmuth, Jorge L Santana Bagur, Olga Mendez, Robert Kalayjian, Kim Whitely, Clyde Crumpacker, Neah Kim, Deborah McMahon, Nancy Mantz, Todd Stroberg, Glenn Sturge, Mitchell Goldman, Deborah O'Connor, Karen Cavanagh, Judith A Aberg, Beverly E Sha, Kristine L Richards, Karen Tashima, Pamela Poethke, Susan L Koletar, Laura Laughlin, Mark Rodriguez, Ge-Youl Kim, Charles E Davis, Barbara Glick, Charles Bradley Hare, Deborah Zeitschel, Henry H Balfour, Kathy Fox, Donna Mildvan, Manuel Revuelt, Nesli Basgoz, Amy Sbrolla, Sandra Valle, Debbie Slamowitz, Ronald Mitsuyasu, Suzette Chafey, Margaret A Fischl, Hector H Bolivar, Jane Reid, Christine Hurley, Ann C Collier, Beck A Royer, Elizabeth Race, Tianna Petersen,
Collapse
|
186
|
Factors associated with collagen deposition in lymphoid tissue in long-term treated HIV-infected patients. AIDS 2010; 24:2029-39. [PMID: 20588162 DOI: 10.1097/qad.0b013e32833c3268] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The factors associated with fibrosis in lymphoid tissue in long-term treated HIV-infected patients and their correlation with immune reconstitution were assessed. METHODS Tonsillar biopsies were performed in seven antiretroviral-naive patients and 29 successfully treated patients (median time on treatment, 61 months). Twenty patients received protease inhibitors-sparing regimens and nine protease inhibitor-containing regimens. Five tonsillar resections of HIV-negative individuals were used as controls. Lymphoid tissue architecture, collagen deposition (fibrosis) and the mean interfollicular CD4(+) cell count per mum were assessed. RESULTS Naive and long-term treated HIV-infected patients had a higher proportion of fibrosis than did HIV-uninfected persons (P < 0.001). Patients with greater collagen deposition showed lower levels of CD4 cells in lymphoid tissue (P = 0.03) and smaller increase in peripheral CD4(+) T cells (r = -0.40, P = 0.05). The factors independently associated with fibrosis in lymphoid tissue were age (P < 0.0001), treated patients with detectable lymphoid tissue viral load when compared with patients with undetectable lymphoid tissue viral load (median 5 vs. 12%, respectively, P = 0.017) and patients receiving a protease inhibitor-sparing vs. a protease inhibitor-containing regimen (median 8 vs. 2.5%, respectively, P = 0.04). CONCLUSION Fibrosis in lymphoid tissue was associated with a poor reconstitution of CD4(+) T cells and long-term antiretroviral therapy did not reverse this abnormality. HIV infection, older age, a detectable level of lymphoid tissue viral load in treated patients and protease inhibitor-sparing regimens seem to favour fibrosis in lymphoid tissue.
Collapse
|
187
|
Tjernlund A, Zhu J, Laing K, Diem K, McDonald D, Vazquez J, Cao J, Ohlen C, McElrath MJ, Picker LJ, Corey L. In situ detection of Gag-specific CD8+ cells in the GI tract of SIV infected Rhesus macaques. Retrovirology 2010; 7:12. [PMID: 20158906 PMCID: PMC2834607 DOI: 10.1186/1742-4690-7-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 02/16/2010] [Indexed: 11/10/2022] Open
Abstract
Background SIV and HIV predominantly replicate in lymphoid tissue, but the study of virus specific CD8+ T cells in intact lymphoid tissue is difficult, as traditional in situ tetramer staining requires fresh tissue. Results In this report, we demonstrate a novel technique using Qdot 655-conjugated peptide-MHC multimers to directly visualize SIV specific cells in cryopreserved tissue biopsies from chronically SIVmac239 infected Rhesus macaques. Qdot 655 multimers showed similar sensitivity and specificity to APC-conjugated tetramers by flow cytometry analysis, but yielded ten-fold higher signal intensity when imaged by fluorescence microscopy. Using this technique, we detected CD8+ T cells which recognize an immunodominant epitope (Gag CM9) in the spleen, lymph nodes, ileum and colon. In all these tissues, the Gag CM9 positive cells were mainly located in the extra follicular T cell zone. In the ileum and colon, we found Gag CM9 positive cells concentrated in Peyer's patches and solitary lymphoid follicles, a pattern of localization not previously described. Conclusions The use of Qdot multimers provide an anatomic and quantitative evaluation of SIV specific CD8+ T cell responses in SIV pathogenesis, and may prove useful to studies of SIV specific CD8+ T cell responses elicited by vaccines and other immunotherapies in the non-human primate model.
Collapse
Affiliation(s)
- Annelie Tjernlund
- Vaccine & Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Hong JJ, Reynolds MR, Mattila TL, Hage A, Watkins DI, Miller CJ, Skinner PJ. Localized populations of CD8 MHC class I tetramer SIV-specific T cells in lymphoid follicles and genital epithelium. PLoS One 2009; 4:e4131. [PMID: 19122815 PMCID: PMC2607009 DOI: 10.1371/journal.pone.0004131] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2008] [Accepted: 12/02/2008] [Indexed: 11/18/2022] Open
Abstract
CD8 T cells play an important role in controlling viral infections. We investigated the in situ localization of simian immunodeficiency virus (SIV)-specific T cells in lymph and genital tissues from SIV-infected macaques using MHC-class I tetramers. The majority of tetramer-binding cells localized in T cell zones and were CD8+. Curiously, small subpopulations of tetramer-binding cells that had little to no surface CD8 were detected in situ both early and late post-infection, and in both vaginally and rectally inoculated macaques. These tetramer+CD8low/− cells were more often localized in apparent B cell follicles relative to T cell zones and more often found near or within the genital epithelium than the submucosa. Cells analyzed by flow cytometry showed similar populations of cells. Further immunohistological characterization revealed small populations of tetramer+CD20− cells inside B cell follicles and that tetramer+ cells did not stain with γδ-TCR nor CD4 antibodies. Negative control tetramer staining indicated that tetramer+CD8low/− cells were not likely NK cells non-specifically binding to MHC tetramers. These findings have important implications for SIV-specific and other antigen-specific T cell function in these specific tissue locations, and suggest a model in which antigen-specific CD8+ T cells down modulate CD8 upon entering B cell follicles or the epithelial layer of tissues, or alternatively a model in which only antigen-specific CD8 T cells that down-modulate CD8 can enter B cell follicles or the epithelium.
Collapse
Affiliation(s)
- Jung Joo Hong
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Matthew R. Reynolds
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Teresa L. Mattila
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Aaron Hage
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - David I. Watkins
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | | | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
189
|
Meditz AL, Schlichtemeier R, Folkvord JM, Givens M, Lesh KC, Ray M, McCarter MD, Connick E. SDF-1alpha is a potent inducer of HIV-1-Specific CD8+ T-cell chemotaxis, but migration of CD8+ T cells is impaired at high viral loads. AIDS Res Hum Retroviruses 2008; 24:977-85. [PMID: 18671480 DOI: 10.1089/aid.2007.0259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiple impairments in HIV-1-specific cytotoxic T cells (CTL) have been reported, but derangements in HIV-1-specific CD8+ T-cell chemotaxis have not been described previously. We assessed migration to SDF-1alpha (stromal cell-derived factor 1-alpha) and CX3CL1 in vitro and expression of cognate receptors, CXCR4 and CX3CR1, by flow cytometry in peripheral blood and lymph node CD8+ T cells from HIV-1-seropositive and -seronegative individuals. Compared with seronegative individuals, percentages of CXCR4+CD8+ T cells were reduced (median, 26% versus 74%, p < 0.001) and percentages of CX3CR1+CD8+ T cells were increased (median, 33% versus 15%, p = 0.03) in seropositive individuals. Robust migration of peripheral blood mononuclear cell (PBMC) CD8+ T cells to SDF-1alpha (1 alphag/ml) was observed in both HIV-1-seropositive (median chemotactic index [CI] 4.9) and -seronegative (median CI 2.8) subjects (p = 0.46). CI to SDF-1alpha was not significantly related to percentage of CXCR4+CD8+ T cells or density of CXCR4, but correlated inversely with plasma HIV-1 RNA concentration (r = -0.82, p = 0.03). Little chemotaxis was observed in response to CX3CL1 and it was unrelated to CX3CR1 expression. Lymph node CD8+ T-cell chemotaxis to SDF-1alpha and CX3CL1 in four subjects demonstrated the same patterns observed in PBMC. HIV-1-specific tetramer-staining CD8+ T cells exhibited chemotaxis of similar magnitude as PBMC CD8+ T cells in a subset of subjects. These data suggest that SDF-1alpha is a potent chemoattractant for HIV-1-specific CTL, but that impairments in migration of HIV-1-specific CTL may exist at high viral loads. Improved understanding of the determinants of CTL localization may provide insight into novel therapies to enhance delivery of CTL to sites of HIV-1 replication.
Collapse
Affiliation(s)
- Amie L. Meditz
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Rick Schlichtemeier
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Joy M. Folkvord
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Monique Givens
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
- Present address: Research and Development, Gambro BCT, Lakewood, Colorado 80215
| | - Kevin C. Lesh
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
- Present address: Lynn Institute of the Rockies, Colorado Springs, Colorado 80909
| | - M.G. Ray
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Martin D. McCarter
- Department of Surgery, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Elizabeth Connick
- Division of Infectious Diseases, Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262
| |
Collapse
|
190
|
Abstract
Since the earliest days of the AIDS epidemic, clinicians and researchers have recognized the importance of lymphoid tissue both in the clinical manifestations of disease and in its pathogenesis. Generalized lymphadenopathy was one of the earliest harbingers of AIDS in the United States and over the past 27 years an increasing body of evidence has implicated the lymphoid organs as central to the pathogenesis of immune deficiency in chronic HIV-1 infection. In this essay, we will review some of the data that have been accumulated and propose a testable model that may reconcile them.
Collapse
Affiliation(s)
- Michael M Lederman
- Center for AIDS Research, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44118, USA.
| | | |
Collapse
|
191
|
|