1
|
Jiang S, Chan CN, Rovira-Clavé X, Chen H, Bai Y, Zhu B, McCaffrey E, Greenwald NF, Liu C, Barlow GL, Weirather JL, Oliveria JP, Nakayama T, Lee IT, Matter MS, Carlisle AE, Philips D, Vazquez G, Mukherjee N, Busman-Sahay K, Nekorchuk M, Terry M, Younger S, Bosse M, Demeter J, Rodig SJ, Tzankov A, Goltsev Y, McIlwain DR, Angelo M, Estes JD, Nolan GP. Combined protein and nucleic acid imaging reveals virus-dependent B cell and macrophage immunosuppression of tissue microenvironments. Immunity 2022; 55:1118-1134.e8. [PMID: 35447093 PMCID: PMC9220319 DOI: 10.1016/j.immuni.2022.03.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 10/13/2021] [Accepted: 03/25/2022] [Indexed: 12/12/2022]
Abstract
Understanding the mechanisms of HIV tissue persistence necessitates the ability to visualize tissue microenvironments where infected cells reside; however, technological barriers limit our ability to dissect the cellular components of these HIV reservoirs. Here, we developed protein and nucleic acid in situ imaging (PANINI) to simultaneously quantify DNA, RNA, and protein levels within these tissue compartments. By coupling PANINI with multiplexed ion beam imaging (MIBI), we measured over 30 parameters simultaneously across archival lymphoid tissues from healthy or simian immunodeficiency virus (SIV)-infected nonhuman primates. PANINI enabled the spatial dissection of cellular phenotypes, functional markers, and viral events resulting from infection. SIV infection induced IL-10 expression in lymphoid B cells, which correlated with local macrophage M2 polarization. This highlights a potential viral mechanism for conditioning an immunosuppressive tissue environment for virion production. The spatial multimodal framework here can be extended to decipher tissue responses in other infectious diseases and tumor biology.
Collapse
Affiliation(s)
- Sizun Jiang
- Department of Pathology, Stanford University, Stanford, CA, USA; Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Chi Ngai Chan
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | | | - Han Chen
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Yunhao Bai
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Bokai Zhu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Erin McCaffrey
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Candace Liu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Graham L Barlow
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Jason L Weirather
- Center of Immuno-Oncology, Dana-Faber Cancer Institute, Boston, MA, USA
| | - John Paul Oliveria
- Department of Pathology, Stanford University, Stanford, CA, USA; Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Tsuguhisa Nakayama
- Department of Pathology, Stanford University, Stanford, CA, USA; Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Ivan T Lee
- Department of Pathology, Stanford University, Stanford, CA, USA; Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthias S Matter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anne E Carlisle
- Center of Immuno-Oncology, Dana-Faber Cancer Institute, Boston, MA, USA
| | - Darci Philips
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Gustavo Vazquez
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Michael Nekorchuk
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Margaret Terry
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Skyler Younger
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Marc Bosse
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Janos Demeter
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham & Women's Hospital, Boston, MA, USA
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Yury Goltsev
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Jacob D Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA; Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA.
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Lu Y, Zhang MX, Pang W, Song TZ, Zheng HY, Tian RR, Zheng YT. Transcription Factor ZNF683 Inhibits SIV/HIV Replication through Regulating IFNγ Secretion of CD8+ T Cells. Viruses 2022; 14:v14040719. [PMID: 35458449 PMCID: PMC9030044 DOI: 10.3390/v14040719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/15/2022] [Accepted: 03/26/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary microbial invasion frequently occurs during AIDS progression in HIV patients. Inflammatory cytokines and other immunoregulatory factors play important roles in this process. We previously established an AIDS model of SIVmac239 infection in northern pig-tailed macaques (NPMs), which were divided into rapid progressor (RP) and slow progressor (SP) groups according to their AIDS progression rates. In this study, we performed 16S rDNA and transcriptome sequencing of the lungs to reveal the molecular mechanism underlying the difference in progression rate between the RPs and SPs. We found that microbial invasion in the RP group was distinct from that in the SP group, showing marker flora of the Family XI, Enterococcus and Ezakiella, and more Lactobacilli. Through pulmonary transcriptome analysis, we found that the transcription factor ZNF683 had higher expression in the SP group than in the RP group. In subsequent functional experiments, we found that ZNF683 increased the proliferation and IFNγ secretion ability of CD8+ T cells, thus decreasing SIV or HIV replication, which may be related to AIDS progression in SIVmac239-infected NPMs. This study helps elucidate the various complexities of disease progression in HIV-1-infected individuals.
Collapse
Affiliation(s)
- Ying Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Ming-Xu Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Wei Pang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Tian-Zhang Song
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Hong-Yi Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Ren-Rong Tian
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (Y.L.); (M.-X.Z.); (W.P.); (T.-Z.S.); (H.-Y.Z.); (R.-R.T.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
- Correspondence: ; Tel.: +86-871-65295684
| |
Collapse
|
3
|
Rodríguez-Castañón JM, Mcnaughton A, Cárdenas-Ochoa A, Fuentes-Romero LL, Viveros-Rogel M, Vergara-Mendoza M, Tello-Mercado AC, Leal-Gutiérrez G, Romero-Carvajal JJ, Cázares-Lara J, Camiro-Zúñiga A, Jaramillo-Jante R, Antuna-Puente B, Galindo-Fraga A, Soto-Ramírez LE, Sierra-Madero JG, Perez-Patrigeon S. Exceptional T CD4 + Recovery Post-antiretroviral Is Linked to a Lower HIV Reservoir with a Specific Immune Differentiation Pattern. AIDS Res Hum Retroviruses 2022; 38:11-21. [PMID: 33779241 DOI: 10.1089/aid.2020.0270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We present a cohort of individuals who reached CD4+ T cell counts of greater than 1,000 cells/mm3 (Hypers) after starting antiretroviral treatment (ART) and compared them with those who reached between 350 and 999 CD4+ T cells/mm3 (Concordants). Demographic data, immune recovery kinetics, T CD4+ subset phenotypes, and integrated HIV DNA were analyzed. Data from individuals living with HIV on their first ART regimen and after 48 months of follow-up were obtained. Immune phenotype by Flow Cytometry analysis on whole blood was performed, cytokines were measured, and integrated HIV-1 DNA was measured by polymerase chain reaction. From a total of 424 individuals, 26 Hypers (6.1%), 314 Concordants (74.1%), and 84 (19.8%) discordants were identified. Hypers had a higher proportion of CD4+-naive (Nv) T cells (37.6 vs. 24.8, p < .05), and a low proportion of CD4+ effector memory T cells (27.9 vs. 39.4, p < .05), with similar results found in CD8+ T cells. Hypers demonstrated a higher percentage of CD4+CD45RA+CD31neg cells with a lower response to interleukin-2 stimulation and a lower integrated HIV-1 DNA/CD4 ratio (1.2 vs. 2.89, p < .05). In Hypers, T cell recovery occurs very early after initiation of ART. Following this initial recovery state, their CD4+ T cell level homeostasis seems to be driven by nonthymic-central-Nv cells. This exceptional recovery is associated with a lower HIV reservoir, which may be related to an increase in noninfected CD4+ T cells. These patients could then be eligible candidates for cure trials.
Collapse
Affiliation(s)
- José Miguel Rodríguez-Castañón
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Andrew Mcnaughton
- Division of Infectious Diseases, Queen's University, Kingston, Canada Kingston, Canada
| | - Ayleen Cárdenas-Ochoa
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Luis León Fuentes-Romero
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Mónica Viveros-Rogel
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Moisés Vergara-Mendoza
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Andrea C. Tello-Mercado
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Graciela Leal-Gutiérrez
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Juan José Romero-Carvajal
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Jonnathan Cázares-Lara
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Antonio Camiro-Zúñiga
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Rocío Jaramillo-Jante
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | | | - Arturo Galindo-Fraga
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Luis E. Soto-Ramírez
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Juan G. Sierra-Madero
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
| | - Santiago Perez-Patrigeon
- Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición salvador Zubirán, Mexico City, Mexico
- Division of Infectious Diseases, Queen's University, Kingston, Canada Kingston, Canada
| |
Collapse
|
4
|
Dabee S, Mkhize NN, Jaspan HB, Lewis D, Gumbi PP, Passmore JAS. Initiation of Antiretroviral Therapy Differentially Influences Genital and Systemic Immune Activation in HIV-Infected Women. AIDS Res Hum Retroviruses 2020; 36:821-830. [PMID: 32524856 DOI: 10.1089/aid.2019.0268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Antiretroviral therapy (ART) has significantly improved the quality of life of HIV-infected individuals: reducing plasma viremia, restoring CD4+ T cell numbers, and correcting imbalances in blood memory T cell subsets. While ART improves immune correlates at mucosal sites, including the lower female genital tract (FGT), ART initiation has been associated with reactivation of common FGT infections. We investigated the effect of ART on immune activation and inflammation in the genital tract. We measured blood and genital T cell activation, proliferation, and immunosenescence (CD38, HLADR, Ki67, CD127, and CD57), and cytokine levels in women on ART for ∼7 years (cross-sectional analysis) or initiating ART (immediately before and 1 month after). Effector memory T cells predominated in blood and FGT during chronic infection, irrespective of ART status. In women initiating ART, 1 month was insufficient for T cell reconstitution, or alterations in T cell subset distribution, despite both plasma and genital viral loads decreasing to undetectable levels in most participants. Initiating ART was accompanied by a decline in plasma IP-10 that correlated with decreased blood CD38 expression in blood (p = .0204) but not in the FGT. The reduction in plasma (but not genital) cytokine levels due to ART initiation was dependent on their concentrations before treatment. While T cell activation decreased significantly in blood (CD4: p = .032; CD8: p = .0137), activation levels remained similar in the genital tract despite 1 month of treatment. Overall, the decrease in cellular activation and inflammation seen in blood with ART initiation was not evident in the FGT.
Collapse
Affiliation(s)
- Smritee Dabee
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | | | - Heather B. Jaspan
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Seattle Childrens Hospital, Seattle, Washington, USA
| | - David Lewis
- Western Sydney Sexual Health Centre, Parramatta, Australia
- Westmead Clinical School and Centre for Infectious Diseases and Microbiology & Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Pamela P. Gumbi
- Department of Biochemistry, University of KwaZulu Natal, Pietermaritzburg, South Africa
| | - Jo-Ann S. Passmore
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- NRF-DST Centre of Excellence in HIV Prevention, Centre for the AIDS Programme of Research in South Africa, University of KwaZulu Natal, Durban, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| |
Collapse
|
5
|
Promoting intestinal lymphatic transport targets a liver-X receptor (LXR) agonist (WAY-252,623) to lymphocytes and enhances immunomodulation. J Control Release 2019; 296:29-39. [PMID: 30611901 DOI: 10.1016/j.jconrel.2019.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/12/2018] [Accepted: 01/02/2019] [Indexed: 12/28/2022]
Abstract
Lymphocytes play a central role in the pathology of a range of chronic conditions such as autoimmune disease, transplant rejection, leukemia, lymphoma HIV/AIDs and cardiometabolic diseases such as atherosclerosis. Current treatments for lymphocyte-associated conditions are incompletely effective and/or complicated by a range of off-target toxicities. One major challenge is poor drug access to lymphocytes via the systemic blood and this may be attributed, at least in part, to the fact that lymphocytes are concentrated within lymph fluid and lymphoid tissues, particularly in gut-associated lymphatics. Here we demonstrate that promoting drug uptake into the intestinal lymphatics with a long chain fatty acid, thereby increasing lymphocyte access, enhances the pharmacodynamic effect of a highly lipophilic liver X receptor (LXR) agonist, WAY-252623, that has been suggested as a potential treatment for atherosclerosis. This has been exemplified by: (1) increased mRNA expression of key markers of LXR activation (ABCA1) and regulatory T cells (Foxp3) in local lymphatic lymphocytes and (2) enhanced numbers of CD4+CD25+Foxp3+ regulatory T cells in the systemic circulation, after administration of a 5-fold lower dose with a lymph directing lipid formulation when compared with a non-lipid containing formulation. These data suggest that combining lipophilic, lymphotropic drug candidates such as WAY-252,623, with lymph-directing long chain lipid based formulations can enhance drug targeting to, and activity on, lymphocytes in lymph and that this effect persists through to the systemic circulation. This presents a promising approach to achieve more selective and effective therapeutic outcomes for the treatment of lymphocyte associated diseases.
Collapse
|
6
|
Extracellular Matrix Proteins Mediate HIV-1 gp120 Interactions with α 4β 7. J Virol 2017; 91:JVI.01005-17. [PMID: 28814519 DOI: 10.1128/jvi.01005-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/09/2017] [Indexed: 01/01/2023] Open
Abstract
Gut-homing α4β7high CD4+ T lymphocytes have been shown to be preferentially targeted by human immunodeficiency virus type 1 (HIV-1) and are implicated in HIV-1 pathogenesis. Previous studies demonstrated that HIV-1 envelope protein gp120 binds and signals through α4β7 and that this likely contributes to the infection of α4β7high T cells and promotes cell-to-cell virus transmission. Structures within the second variable loop (V2) of gp120, including the tripeptide motif LDV/I, are thought to mediate gp120-α4β7 binding. However, lack of α4β7 binding has been reported in gp120 proteins containing LDV/I, and the precise determinants of gp120-α4β7 binding are not fully defined. In this work, we report the novel finding that fibronectins mediate indirect gp120-α4β7 interactions. We show that Chinese hamster ovary (CHO) cells used to express recombinant gp120 produced fibronectins and other extracellular matrix proteins that copurified with gp120. CHO cell fibronectins were able to mediate the binding of a diverse panel of gp120 proteins to α4β7 in an in vitro cell binding assay. The V2 loop was not required for fibronectin-mediated binding of gp120 to α4β7, nor did V2-specific antibodies block this interaction. Removal of fibronectin through anion-exchange chromatography abrogated V2-independent gp120-α4β7 binding. Additionally, we showed a recombinant human fibronectin fragment mediated gp120-α4β7 interactions similarly to CHO cell fibronectin. These findings provide an explanation for the apparently contradictory observations regarding the gp120-α4β7 interaction and offer new insights into the potential role of fibronectin and other extracellular matrix proteins in HIV-1 biology.IMPORTANCE Immune tissues within the gut are severely damaged by HIV-1, and this plays an important role in the development of AIDS. Integrin α4β7 plays a major role in the trafficking of lymphocytes, including CD4+ T cells, into gut lymphoid tissues. Previous reports indicate that some HIV-1 gp120 envelope proteins bind to and signal through α4β7, which may help explain the preferential infection of gut CD4+ T cells. In this study, we demonstrate that extracellular matrix proteins can mediate interactions between gp120 and α4β7 This suggests that the extracellular matrix may be an important mediator of HIV-1 interaction with α4β7-expressing cells. These findings provide new insight into the nature of HIV-1-α4β7 interactions and how these interactions may represent targets for therapeutic intervention.
Collapse
|
7
|
Kijak GH, Sanders-Buell E, Chenine AL, Eller MA, Goonetilleke N, Thomas R, Leviyang S, Harbolick EA, Bose M, Pham P, Oropeza C, Poltavee K, O’Sullivan AM, Billings E, Merbah M, Costanzo MC, Warren JA, Slike B, Li H, Peachman KK, Fischer W, Gao F, Cicala C, Arthos J, Eller LA, O’Connell RJ, Sinei S, Maganga L, Kibuuka H, Nitayaphan S, Rao M, Marovich MA, Krebs SJ, Rolland M, Korber BT, Shaw GM, Michael NL, Robb ML, Tovanabutra S, Kim JH. Rare HIV-1 transmitted/founder lineages identified by deep viral sequencing contribute to rapid shifts in dominant quasispecies during acute and early infection. PLoS Pathog 2017; 13:e1006510. [PMID: 28759651 PMCID: PMC5552316 DOI: 10.1371/journal.ppat.1006510] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 08/10/2017] [Accepted: 07/03/2017] [Indexed: 01/12/2023] Open
Abstract
In order to inform the rational design of HIV-1 preventive and cure interventions it is critical to understand the events occurring during acute HIV-1 infection (AHI). Using viral deep sequencing on six participants from the early capture acute infection RV217 cohort, we have studied HIV-1 evolution in plasma collected twice weekly during the first weeks following the advent of viremia. The analysis of infections established by multiple transmitted/founder (T/F) viruses revealed novel viral profiles that included: a) the low-level persistence of minor T/F variants, b) the rapid replacement of the major T/F by a minor T/F, and c) an initial expansion of the minor T/F followed by a quick collapse of the same minor T/F to low frequency. In most participants, cytotoxic T-lymphocyte (CTL) escape was first detected at the end of peak viremia downslope, proceeded at higher rates than previously measured in HIV-1 infection, and usually occurred through the exploration of multiple mutational pathways within an epitope. The rapid emergence of CTL escape variants suggests a strong and early CTL response. Minor T/F viral strains can contribute to rapid and varied profiles of HIV-1 quasispecies evolution during AHI. Overall, our results demonstrate that early, deep, and frequent sampling is needed to investigate viral/host interaction during AHI, which could help identify prerequisites for prevention and cure of HIV-1 infection.
Collapse
Affiliation(s)
- Gustavo H. Kijak
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
- * E-mail:
| | - Eric Sanders-Buell
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Agnes-Laurence Chenine
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Michael A. Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Nilu Goonetilleke
- School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Rasmi Thomas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Sivan Leviyang
- Department of Mathematics and Statistics, Georgetown University, Washington, DC, United States of America
| | - Elizabeth A. Harbolick
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Meera Bose
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Phuc Pham
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Celina Oropeza
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Kultida Poltavee
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Anne Marie O’Sullivan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Erik Billings
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Melanie Merbah
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Margaret C. Costanzo
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Joanna A. Warren
- School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Bonnie Slike
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Hui Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Kristina K. Peachman
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Will Fischer
- Theoretical Biology, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Feng Gao
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
| | - Claudia Cicala
- Laboratory of Immunoregulation National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - James Arthos
- Laboratory of Immunoregulation National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Leigh A. Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | | | | | | | - Hannah Kibuuka
- Makerere University-Walter Reed Project, Kampala, Uganda
| | | | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Mary A. Marovich
- Vaccine Research Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States of America
| | - Shelly J. Krebs
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Morgane Rolland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Bette T. Korber
- Theoretical Biology, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - George M. Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Nelson L. Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Merlin L. Robb
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Sodsai Tovanabutra
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Jerome H. Kim
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| |
Collapse
|
8
|
Feder AF, Kline C, Polacino P, Cottrell M, Kashuba ADM, Keele BF, Hu SL, Petrov DA, Pennings PS, Ambrose Z. A spatio-temporal assessment of simian/human immunodeficiency virus (SHIV) evolution reveals a highly dynamic process within the host. PLoS Pathog 2017; 13:e1006358. [PMID: 28542550 PMCID: PMC5444849 DOI: 10.1371/journal.ppat.1006358] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/17/2017] [Indexed: 12/25/2022] Open
Abstract
The process by which drug-resistant HIV-1 arises and spreads spatially within an infected individual is poorly understood. Studies have found variable results relating how HIV-1 in the blood differs from virus sampled in tissues, offering conflicting findings about whether HIV-1 throughout the body is homogeneously distributed. However, most of these studies sample only two compartments and few have data from multiple time points. To directly measure how drug resistance spreads within a host and to assess how spatial structure impacts its emergence, we examined serial sequences from four macaques infected with RT-SHIVmne027, a simian immunodeficiency virus encoding HIV-1 reverse transcriptase (RT), and treated with RT inhibitors. Both viral DNA and RNA (vDNA and vRNA) were isolated from the blood (including plasma and peripheral blood mononuclear cells), lymph nodes, gut, and vagina at a median of four time points and RT was characterized via single-genome sequencing. The resulting sequences reveal a dynamic system in which vRNA rapidly acquires drug resistance concomitantly across compartments through multiple independent mutations. Fast migration results in the same viral genotypes present across compartments, but not so fast as to equilibrate their frequencies immediately. The blood and lymph nodes were found to be compartmentalized rarely, while both the blood and lymph node were more frequently different from mucosal tissues. This study suggests that even oft-sampled blood does not fully capture the viral dynamics in other parts of the body, especially the gut where vRNA turnover was faster than the plasma and vDNA retained fewer wild-type viruses than other sampled compartments. Our findings of transient compartmentalization across multiple tissues may help explain the varied results of previous compartmentalization studies in HIV-1.
Collapse
Affiliation(s)
- Alison F. Feder
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Christopher Kline
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Patricia Polacino
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Mackenzie Cottrell
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Angela D. M. Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Shiu-Lok Hu
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Dmitri A. Petrov
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Pleuni S. Pennings
- Department of Biology, San Francisco State University, San Francisco, CA, United States
| | - Zandrea Ambrose
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Jaworski JP, Vendrell A, Chiavenna SM. Neutralizing Monoclonal Antibodies to Fight HIV-1: On the Threshold of Success. Front Immunol 2017; 7:661. [PMID: 28123384 PMCID: PMC5225137 DOI: 10.3389/fimmu.2016.00661] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022] Open
Abstract
Anti-human immunodeficiency virus type-1 (anti-HIV-1) neutralizing monoclonal antibodies are broadening the spectrum of pre- and post-exposure treatment against HIV-1. A better understanding of how these antibodies develop and interact with particular regions of the viral envelope protein is guiding a more rational structure-based immunogen design. The aim of this article is to review the most recent advances in the field, from the development of these particular antibodies during natural HIV-1 infection, to their role preventing infection, boosting endogenous immune responses and clearing both free viral particles and persistently infected cells.
Collapse
Affiliation(s)
- Juan Pablo Jaworski
- National Scientific and Technical Research Council, Buenos Aires, Argentina; Institute of Virology, National Institute of Agricultural Technology, Castelar, Buenos Aires, Argentina
| | - Alejandrina Vendrell
- Pharmacological and Botanical Study Center, School of Medicine, University of Buenos Aires , Buenos Aires , Argentina
| | | |
Collapse
|
10
|
Immune activation in the central nervous system throughout the course of HIV infection. Curr Opin HIV AIDS 2016; 11:226-33. [PMID: 26760827 DOI: 10.1097/coh.0000000000000243] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Robust and dynamic innate and adaptive responses characterize the acute central nervous system (CNS) response to HIV and other viral infections. In a state of chronic infection or viral latency, persistent immune activation associates with abnormality in the CNS. Understanding this process is critical, as immune-mediated abnormality in nonrenewable CNS cells may result in long-term neurologic sequelae for HIV-infected individuals. RECENT FINDINGS In humans, immune activation is reduced by suppressive combination antiretroviral therapy, but persists at abnormally elevated levels on treatment. CNS immune activation is initiated in acute infection and progressively increases until combination antiretroviral therapy is started. Newly identified characteristics of the CNS immune surveillance network include features of homeostasis and function of brain microglial cells, lymphatic drainage from CNS to cervical lymph nodes, and cells in cerebrospinal fluid associated with neurocognitive impairment. SUMMARY More research is required to determine whether early intervention to reduce infection limits the immunopathology established by sustained immune responses that ultimately fail to resolve infection, and to unravel mechanisms of persistent immune activation during treated HIV so that strategies can be developed to therapeutically protect the brain.
Collapse
|
11
|
Lama JR, Karuna ST, Grant SP, Swann EM, Ganoza C, Segura P, Montano SM, Lacherre M, De Rosa SC, Buchbinder S, Sanchez J, McElrath MJ, Lemos MP. Transient Peripheral Immune Activation follows Elective Sigmoidoscopy or Circumcision in a Cohort Study of MSM at Risk of HIV Infection. PLoS One 2016; 11:e0160487. [PMID: 27536938 PMCID: PMC4990246 DOI: 10.1371/journal.pone.0160487] [Citation(s) in RCA: 6] [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: 03/22/2016] [Accepted: 07/20/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Rectal and genital sampling in HIV prevention trials permits assessments at the site of HIV entry. Yet the safety and acceptability of circumcision and sigmoidoscopy (and associated abstinence recommendations) are unknown in uncircumcised men who have sex with men (MSM) at high risk of HIV infection. METHODS Twenty-nine HIV-seronegative high-risk Peruvian MSM agreed to elective sigmoidoscopy biopsy collections (weeks 2 and 27) and circumcision (week 4) in a 28-week cohort study designed to mimic an HIV vaccine study mucosal collection protocol. We monitored adherence to abstinence recommendations, procedure-related complications, HIV infections, peripheral immune activation, and retention. RESULTS Twenty-three (79.3%) underwent a first sigmoidoscopy, 21 (72.4%) were circumcised, and 16 (55.2%) completed a second sigmoidoscopy during the study period. All who underwent procedures completed the associated follow-up safety visits. Those completing the procedures reported they were well tolerated, and complication rates were similar to those reported in the literature. Immune activation was detected during the healing period (1 week post-sigmoidoscopy, 6 weeks post-circumcision), including increases in CCR5+CD4+T cells and α4β7+CD4+T cells. Most participants adhered to post-circumcision abstinence recommendations whereas reduced adherence occurred post-sigmoidoscopy. CONCLUSION Rectosigmoid mucosal and genital tissue collections were safe in high-risk MSM. Although the clinical implications of the post-procedure increase in peripheral immune activation markers are unknown, they reinforce the need to provide ongoing risk reduction counseling and support for post-procedure abstinence recommendations. Future HIV vaccine studies should also consider the effects of mucosal and tissue collections on peripheral blood endpoints in trial design and analysis. TRIAL REGISTRATION ClinicalTrials.gov NCT02630082.
Collapse
Affiliation(s)
| | - Shelly T. Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Shannon P. Grant
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Edith M. Swann
- Vaccine Clinical Research Branch, Division of AIDS, National Institutes of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, Maryland, United States of America
| | | | | | | | | | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Susan Buchbinder
- San Francisco Department of Health, San Francisco, California, United States of America
| | - Jorge Sanchez
- Asociacion Civil Impacta Salud y Educacion, Lima, Peru
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Maria P. Lemos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | | |
Collapse
|
12
|
DeGottardi MQ, Okoye AA, Vaidya M, Talla A, Konfe AL, Reyes MD, Clock JA, Duell DM, Legasse AW, Sabnis A, Park BS, Axthelm MK, Estes JD, Reiman KA, Sekaly RP, Picker LJ. Effect of Anti-IL-15 Administration on T Cell and NK Cell Homeostasis in Rhesus Macaques. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:1183-98. [PMID: 27430715 PMCID: PMC4976006 DOI: 10.4049/jimmunol.1600065] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023]
Abstract
IL-15 has been implicated as a key regulator of T and NK cell homeostasis in multiple systems; however, its specific role in maintaining peripheral T and NK cell populations relative to other γ-chain (γc) cytokines has not been fully defined in primates. In this article, we address this question by determining the effect of IL-15 inhibition with a rhesusized anti-IL-15 mAb on T and NK cell dynamics in rhesus macaques. Strikingly, anti-IL-15 treatment resulted in rapid depletion of NK cells and both CD4(+) and CD8(+) effector memory T cells (TEM) in blood and tissues, with little to no effect on naive or central memory T cells. Importantly, whereas depletion of NK cells was nearly complete and maintained as long as anti-IL-15 treatment was given, TEM depletion was countered by the onset of massive TEM proliferation, which almost completely restored circulating TEM numbers. Tissue TEM, however, remained significantly reduced, and most TEM maintained very high turnover throughout anti-IL-15 treatment. In the presence of IL-15 inhibition, TEM became increasingly more sensitive to IL-7 stimulation in vivo, and transcriptional analysis of TEM in IL-15-inhibited monkeys revealed engagement of the JAK/STAT signaling pathway, suggesting alternative γc cytokine signaling may support TEM homeostasis in the absence of IL-15. Thus, IL-15 plays a major role in peripheral maintenance of NK cells and TEM However, whereas most NK cell populations collapse in the absence of IL-15, TEM can be maintained in the face of IL-15 inhibition by the activity of other homeostatic regulators, most likely IL-7.
Collapse
Affiliation(s)
- Maren Q DeGottardi
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Afam A Okoye
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Mukta Vaidya
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Aarthi Talla
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Audrie L Konfe
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Matthew D Reyes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Joseph A Clock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Derick M Duell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Alfred W Legasse
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Amit Sabnis
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Byung S Park
- Division of Biostatistics, Department of Public Health and Preventative Medicine, Oregon Health & Science University, Portland, OR 97239
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD 21702; and
| | - Keith A Reiman
- MassBiologics, University of Massachusetts Medical School, Boston, MA 02126
| | | | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006;
| |
Collapse
|
13
|
Neuropsychological Impairment in Acute HIV and the Effect of Immediate Antiretroviral Therapy. J Acquir Immune Defic Syndr 2016; 70:393-9. [PMID: 26509933 DOI: 10.1097/qai.0000000000000746] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate neuropsychological performance (NP) during acute HIV infection (AHI) before and after combination antiretroviral therapy (cART). DESIGN Prospective study of Thai AHI participants examined at 3 and 6 months after initiation of cART. METHODS Thirty-six AHI participants were evaluated pre-cART at median 19 days since HIV exposure and 3 and 6 months after cART with the Grooved Pegboard test, Color Trails 1 & 2 (CT1, CT2), and Trail Making Test A. Raw scores were standardized to 251 age- and education-matched HIV-uninfected Thais. To account for learning effects, change in NP performance was compared with that of controls at 6 months. Analyses included multivariable regression, nonparametric repeated measures analysis of variance, and Mann-Whitney U test. RESULTS Baseline NP scores for the AHI group were within normal range (z-scores range: -0.26 to -0.13). NP performance improved on CT1, CT2, and Trail Making Test A in the initial 3 months (P < 0.01) with no significant change during the last 3 months. Only improvement in CT1 was greater than that seen in controls at 6 months (P = 0.018). Participants who performed >1 SD below normative means on ≥2 tests (n = 8) exhibited higher baseline cerebrospinal fluid HIV RNA (P = 0.047) and had no improvement after cART. CONCLUSIONS Most AHI individuals had normal NP performance, and early cART slightly improved their psychomotor function. However, approximately 25% had impaired NP performance, which correlated with higher cerebrospinal fluid HIV RNA, and these abnormalities were not reversed by early cART possibly indicating limited reversibility of cognitive impairment in a subset of AHI individuals.
Collapse
|
14
|
Terahara K, Ishige M, Ikeno S, Okada S, Kobayashi-Ishihara M, Ato M, Tsunetsugu-Yokota Y. Humanized mice dually challenged with R5 and X4 HIV-1 show preferential R5 viremia and restricted X4 infection of CCR5(+)CD4(+) T cells. Microbes Infect 2015; 17:378-86. [PMID: 25839960 DOI: 10.1016/j.micinf.2015.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 11/30/2022]
Abstract
CCR5-tropic (R5) immunodeficiency virus type 1 (HIV-1) strains are highly transmissible during the early stage of infection in humans, whereas CXCR4-tropic (X4) strains are less transmissible. This study aimed to explore the basis for early phase R5 and X4 HIV-1 infection in vivo by using humanized mice dually challenged with R5 HIV-1NLAD8-D harboring DsRed and X4 HIV-1(NL-E) harboring EGFP. Whereas R5 HIV-1 replicated well, X4 HIV-1 caused only transient viremia with variable kinetics; however, this was distinct from the low level but persistent viremia observed in mice challenged with X4 HIV-1 alone. Flow cytometric analysis of HIV-1-infected cells revealed that X4 HIV-1 infection of CCR5(+)CD4(+) T cells was significantly suppressed in the presence of R5 HIV-1. X4 HIV-1 was more cytopathic than R5 HIV-1; however, this was not the cause of restricted X4 HIV-1 infection because there were no significant differences in the mortality rates of CCR5(+) and CCR5(-) cells within the X4 HIV-1-infected cell populations. Taken together, these results suggest that restricted infection of CCR5(+)CD4(+) T cells by X4 HIV-1 (occurring via a still-to-be-identified mechanism) might contribute to the preferential transmission of R5 HIV-1 during the early phase of infection.
Collapse
Affiliation(s)
- Kazutaka Terahara
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Masayuki Ishige
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | - Shota Ikeno
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; Cooperative Major in Advanced Health Science, Tokyo University of Agriculture and Technology/Waseda University Graduate School of Collaborative Education Curriculum, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | - Mie Kobayashi-Ishihara
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; Japan Foundation for AIDS Prevention, 1-3-12 Misakimachi, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Manabu Ato
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Yasuko Tsunetsugu-Yokota
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; Department of Medical Technology, School of Human Sciences, Tokyo University of Technology, 5-23-22 Nishikamata, Ota-ku, Tokyo 144-8535, Japan.
| |
Collapse
|
15
|
Santangelo PJ, Rogers KA, Zurla C, Blanchard EL, Gumber S, Strait K, Connor-Stroud F, Schuster DM, Amancha PK, Hong JJ, Byrareddy SN, Hoxie JA, Vidakovic B, Ansari AA, Hunter E, Villinger F. Whole-body immunoPET reveals active SIV dynamics in viremic and antiretroviral therapy-treated macaques. Nat Methods 2015; 12:427-32. [PMID: 25751144 DOI: 10.1038/nmeth.3320] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 02/07/2015] [Indexed: 11/09/2022]
Abstract
The detection of viral dynamics and localization in the context of controlled HIV infection remains a challenge and is limited to blood and biopsies. We developed a method to capture total-body simian immunodeficiency virus (SIV) replication using immunoPET (antibody-targeted positron emission tomography). The administration of a poly(ethylene glycol)-modified, (64)Cu-labeled SIV Gp120-specific antibody led to readily detectable signals in the gastrointestinal and respiratory tract, lymphoid tissues and reproductive organs of viremic monkeys. Viral signals were reduced in aviremic antiretroviral-treated monkeys but detectable in colon, select lymph nodes, small bowel, nasal turbinates, the genital tract and lung. In elite controllers, virus was detected primarily in foci in the small bowel, select lymphoid areas and the male reproductive tract, as confirmed by quantitative reverse-transcription PCR (qRT-PCR) and immunohistochemistry. This real-time, in vivo viral imaging method has broad applications to the study of immunodeficiency virus pathogenesis, drug and vaccine development, and the potential for clinical translation.
Collapse
Affiliation(s)
- Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Kenneth A Rogers
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Chiara Zurla
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Emmeline L Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Sanjeev Gumber
- 1] Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA. [2] Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Karen Strait
- Division of Veterinary Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Fawn Connor-Stroud
- Division of Veterinary Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - David M Schuster
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Praveen K Amancha
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Jung Joo Hong
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Siddappa N Byrareddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - James A Hoxie
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Brani Vidakovic
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Aftab A Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eric Hunter
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Francois Villinger
- 1] Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA. [2] Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
16
|
HIV enteropathy and aging: gastrointestinal immunity, mucosal epithelial barrier, and microbial translocation. Curr Opin HIV AIDS 2015; 9:309-16. [PMID: 24871087 DOI: 10.1097/coh.0000000000000066] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Despite decreases in morbidity and mortality as a result of antiretroviral therapy, gastrointestinal dysfunction remains common in HIV infection. Treated patients are at risk for complications of 'premature' aging, such as cardiovascular disease, osteopenia, neurocognitive decline, malignancies, and frailty. This review summarizes recent observations in this field. RECENT FINDINGS Mucosal CD4 lymphocytes, especially Th17 cells, are depleted in acute HIV and simian immune deficiency virus (SIV) infections, although other cell types also are affected. Reconstitution during therapy often is incomplete, especially in mucosa. Mucosal barrier function is affected by both HIV infection and aging and includes paracellular transport via tight junctions and uptake through areas of apoptosis; other factors may affect systemic antigen exposure. The resultant microbial translocation is associated with systemic immune activation in HIV and SIV infections. There is evidence of immune activation and microbial translocation in the elderly. The immune phenotypes of immunosenescence in HIV infection and aging appear similar. There are several targets for intervention; blockage of residual mucosal virus replication, preventing antigen uptake, modulating the microbiome, improving T cell recovery, combining therapies aimed at mucosal integrity, augmenting mucosal immunity, and managing traditional risk factors for premature aging in the general population. SUMMARY Aging may interact with HIV enteropathy to enhance microbial translocation and immune activation.
Collapse
|
17
|
Persistent inflammation in HIV infection: Established concepts, new perspectives. Immunol Lett 2014; 161:184-8. [DOI: 10.1016/j.imlet.2014.01.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 01/12/2014] [Indexed: 11/20/2022]
|
18
|
Furuya AKM, Sharifi HJ, de Noronha CMC. The Curious Case of Type I IFN and MxA: Tipping the Immune Balance in AIDS. Front Immunol 2014; 5:419. [PMID: 25228901 PMCID: PMC4151092 DOI: 10.3389/fimmu.2014.00419] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 08/18/2014] [Indexed: 01/30/2023] Open
Affiliation(s)
| | - Hamayun J Sharifi
- Albany Medical Center, Center for Immunology and Microbial Disease , Albany, NY , USA
| | - Carlos M C de Noronha
- Albany Medical Center, Center for Immunology and Microbial Disease , Albany, NY , USA
| |
Collapse
|
19
|
Neutralising properties of peptides derived from CXCR4 extracellular loops towards CXCL12 binding and HIV-1 infection. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1031-41. [PMID: 24480462 DOI: 10.1016/j.bbamcr.2014.01.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 12/23/2013] [Accepted: 01/17/2014] [Indexed: 01/01/2023]
Abstract
The chemokine receptor CXCR4 interacts with a single endogenous chemokine, CXCL12, and regulates a wide variety of physiological and pathological processes including inflammation and metastasis development. CXCR4 also binds the HIV-1 envelope glycoprotein, gp120, resulting in viral entry into host cells. Therefore, CXCR4 and its ligands represent valuable drug targets. In this study, we investigated the inhibitory properties of synthetic peptides derived from CXCR4 extracellular loops (ECL1-X4, ECL2-X4 and ECL3-X4) towards HIV-1 infection and CXCL12-mediated receptor activation. Among these peptides, ECL1-X4 displayed anti-HIV-1 activity against X4, R5/X4 and R5 viruses (IC50=24 to 76μM) in cell viability assay without impairing physiological CXCR4-CXCL12 signalling. In contrast, ECL2-X4 only inhibited X4 and R5/X4 strains, interfering with HIV-entry into cells. At the same time, ECL2-X4 strongly and specifically interacted with CXCL12, blocking its binding to CXCR4 and its second receptor, CXCR7 (IC50=20 and 100μM). Further analysis using mutated and truncated peptides showed that ECL2 of CXCR4 forms multiple contacts with the gp120 protein and the N-terminus of CXCL12. Chemokine neutralisation was mainly driven by four aspartates and the C-terminal residues of ECL2-X4. These results demonstrate that ECL2 represents an important structural determinant in CXCR4 activation. We identified the putative site for the binding of CXCL12 N-terminus and provided new structural elements to explain the recognition of gp120 and dimeric CXCR4 ligands.
Collapse
|
20
|
Okoye AA, Picker LJ. CD4(+) T-cell depletion in HIV infection: mechanisms of immunological failure. Immunol Rev 2014; 254:54-64. [PMID: 23772614 DOI: 10.1111/imr.12066] [Citation(s) in RCA: 346] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The hallmark of acquired immunodeficiency syndrome (AIDS) pathogenesis is a progressive depletion of CD4(+) T-cell populations in close association with progressive impairment of cellular immunity and increasing susceptibility to opportunistic infections (OI). Disease progression in untreated human immunodeficiency virus (HIV) infection can take many years, and it was originally hypothesized to be a consequence of slow, viral-mediated CD4(+) T-cell destruction. However, massive CD4(+) memory T-cell destruction is now known to occur quite early in infection, almost always without overt immunodeficiency. In most individuals, this initial destruction is countered by CD4(+) memory T-cell regeneration that preserves CD4(+) T-cell numbers and functions above the threshold associated with overt immunodeficiency. This regeneration, which occurs in the setting of chronic immune activation and immune dysregulation does not, however, restore all functionally important CD4(+) T-cell populations and is not stable over the long term. Ultimately, CD4(+) memory T-cell homeostasis fails and critical effector populations decline below the level necessary to prevent OI. Thus, the onset of overt immune deficiency appears to be intimately linked with CD4(+) memory T-cell dynamics and reflects the complex interplay of direct viral cytopathogenicity and the indirect effects of persistent immune activation on CD4(+) memory T-cell proliferation, differentiation, and survival.
Collapse
Affiliation(s)
- Afam A Okoye
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | | |
Collapse
|
21
|
Abstract
Recent advances in the immunology, pathogenesis, and prevention of human immunodeficiency virus (HIV) infection continue to reveal clues to the mechanisms involved in the progressive immunodeficiency attributed to infection, but more importantly have shed light on the correlates of immunity to infection and disease progression. HIV selectively infects, eliminates, and/or dysregulates several key cells of the human immune system, thwarting multiple arms of the host immune response, and inflicting severe damage to mucosal barriers, resulting in tissue infiltration of 'symbiotic' intestinal bacteria and viruses that essentially become opportunistic infections promoting systemic immune activation. This leads to activation and recruitment or more target cells for perpetuating HIV infection, resulting in persistent, high-level viral replication in lymphoid tissues, rapid evolution of resistant strains, and continued evasion of immune responses. However, vaccine studies and studies of spontaneous controllers are finally providing correlates of immunity from protection and disease progression, including virus-specific CD4(+) T-cell responses, binding anti-bodies, innate immune responses, and generation of antibodies with potent antibody-dependent cell-mediated cytotoxicity activity. Emerging correlates of immunity indicate that prevention of HIV infection may be possible through effective vaccine strategies that protect and stimulate key regulatory cells and immune responses in susceptible hosts. Furthermore, immune therapies specifically directed toward boosting specific aspects of the immune system may eventually lead to a cure for HIV-infected patients.
Collapse
Affiliation(s)
- Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | | | | |
Collapse
|
22
|
Boasso A. Type I Interferon at the Interface of Antiviral Immunity and Immune Regulation: The Curious Case of HIV-1. SCIENTIFICA 2013; 2013:580968. [PMID: 24455433 PMCID: PMC3885208 DOI: 10.1155/2013/580968] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 12/10/2013] [Indexed: 06/03/2023]
Abstract
Type I interferon (IFN-I) play a critical role in the innate immune response against viral infections. They actively participate in antiviral immunity by inducing molecular mechanisms of viral restriction and by limiting the spread of the infection, but they also orchestrate the initial phases of the adaptive immune response and influence the quality of T cell immunity. During infection with the human immunodeficiency virus type 1 (HIV-1), the production of and response to IFN-I may be severely altered by the lymphotropic nature of the virus. In this review I consider the different aspects of virus sensing, IFN-I production, signalling, and effects on target cells, with a particular focus on the alterations observed following HIV-1 infection.
Collapse
Affiliation(s)
- Adriano Boasso
- Immunology Section, Chelsea and Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK
| |
Collapse
|
23
|
Stebbings R, Li B, Lorin C, Koutsoukos M, Février M, Mee ET, Page M, Almond N, Tangy F, Voss G. Immunogenicity of a recombinant measles HIV-1 subtype C vaccine. Vaccine 2013; 31:6079-86. [PMID: 24161574 DOI: 10.1016/j.vaccine.2013.09.072] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/09/2013] [Accepted: 09/30/2013] [Indexed: 12/21/2022]
Abstract
The HIV epidemic is greatest in Sub-Saharan Africa and India where HIV-1 subtype C is predominant. To control the spread of HIV in these parts of the world a preventive HIV-1 subtype C vaccine is urgently required. Here we report the immunogenicity of a candidate HIV-1 subtype C vaccine delivered by a recombinant measles vector carrying an insert encoding HIV-1 subtype C Gag, RT and Nef (MV1-F4), in MHC-typed non-human primates. HIV-1 specific cytokine secreting CD4+ and CD8+ T cell responses were detected in 15 out of 16 vaccinees. These HIV-specific T cell responses persisted in lymphoid tissues. Anti-HIV-1 antibody responses were detected in 15 out of 16 vaccinees and titres were boosted by a second immunisation carried out 84 days later. These findings support further exploration of the MV1-F4 vector as a candidate HIV-1 subtype C vaccine or as part of a wider vaccine strategy.
Collapse
Affiliation(s)
- Richard Stebbings
- Division of Biotherapeutics, NIBSC, Potters Bar, Hertfordshire EN6 3QG, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The spectrum of HIV-associated neurocognitive disorder (HAND) has been dramatically altered in the setting of widely available effective antiretroviral therapy (ART). Once culminating in dementia in many individuals infected with HIV, HAND now typically manifests as more subtle, though still morbid, forms of cognitive impairment in persons surviving long-term with treated HIV infection. Despite the substantial improvement in severity of this disorder, the fact that neurologic injury persists despite ART remains a challenge to the community of patients, providers and investigators aiming to optimize quality of life for those living with HIV. Cognitive dysfunction in treated HIV may reflect early irreversible CNS injury accrued before ART is typically initiated, ongoing low-level CNS infection and progressive injury in the setting of ART, or comborbidities including effects of treatment which may confound the beneficial reduction in viral replication and immune activation effected by ART.
Collapse
|
25
|
Lederman MM, Funderburg NT, Sekaly RP, Klatt NR, Hunt PW. Residual immune dysregulation syndrome in treated HIV infection. Adv Immunol 2013; 119:51-83. [PMID: 23886064 DOI: 10.1016/b978-0-12-407707-2.00002-3] [Citation(s) in RCA: 259] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antiretroviral therapy has revolutionized the course of HIV infection, improving immune function and decreasing dramatically the mortality and morbidity due to the opportunistic complications of the disease. Nonetheless, even with sustained suppression of HIV replication, many HIV-infected persons experience a syndrome characterized by increased T cell activation and evidence of heightened inflammation and coagulation. This residual immune dysregulation syndrome or RIDS is more common in persons who fail to increase circulating CD4+ T cells to normal levels and in several epidemiologic studies it has been associated with increased morbidity and mortality. These morbid and fatal events are not the typical opportunistic infections and malignancies seen in the early AIDS era but rather comprise a spectrum of cardiovascular events, liver disease, metabolic disorders, kidney disease, bone disease, and a spectrum of malignant complications distinguishable from the opportunistic malignancies that characterized the earlier days of the AIDS epidemic. While immune activation, inflammation, and coagulopathy are characteristic of untreated HIV infection and improve with drug-induced control of HIV replication, the drivers of RIDS in treated HIV infection are incompletely understood. And while inflammation, immune activation, and coagulopathy are more common in treated persons who fail to restore circulating CD4+ T cells, it is not entirely clear how these two phenomena are linked.
Collapse
Affiliation(s)
- Michael M Lederman
- Division of Infectious Diseases and Center for AIDS Research, Case Western Reserve University and University Hospitals/Case Medical Center, Cleveland, Ohio, USA.
| | | | | | | | | |
Collapse
|
26
|
Estimating the contribution of the gut to plasma viral load in early SIV infection. Retrovirology 2013; 10:105. [PMID: 24119218 PMCID: PMC3854614 DOI: 10.1186/1742-4690-10-105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 09/22/2013] [Indexed: 11/10/2022] Open
Abstract
Background There is significant debate about whether the gut plays a major role in viral replication and pathology in HIV infection. Here we aimed to estimate the contribution of the gut to the total virus observed in plasma, by comparing the frequency of different viral mutants in plasma and gut in SIV infection. Results We found that the maximum contribution of gut to plasma viral load estimated from rectal biopsy at day 28 post-infection had a median of 10%. The estimated values for individual animals ranged from nearly 100% to <3% in 4/14 animals. Importantly, these are maximum estimates, so that a value of 90%, for example, means that the real contribution may be anything between 0 and 90%, just not higher than 90%. We also studied the contribution of gut at the peak of plasma viral load (day 14). However, since there was very little escape in most animals at this time point, we could only estimate the maximal contribution of gut in 4 animals, in two of which it was <15%. Conclusions The role of the gut in HIV is a controversial area, with many suggesting that it plays a dominant role in driving early infection. Our analysis suggests that, at least by day 28 post-infection, the gut is not contributing greatly to the plasma viral load.
Collapse
|
27
|
Abbas W, Herbein G. T-Cell Signaling in HIV-1 Infection. Open Virol J 2013; 7:57-71. [PMID: 23986795 PMCID: PMC3751038 DOI: 10.2174/1874357920130621001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 05/31/2013] [Accepted: 06/04/2013] [Indexed: 12/20/2022] Open
Abstract
HIV exploits the T-cell signaling network to gain access to downstream cellular components, which serves as effective tools to break the cellular barriers. Multiple host factors and their interaction with viral proteins contribute to the complexity of HIV-1 pathogenesis and disease progression. HIV-1 proteins gp120, Nef, Tat and Vpr alter the T-cell signaling pathways by activating multiple transcription factors including NF-ĸB, Sp1 and AP-1. HIV-1 evades the immune system by developing a multi-pronged strategy. Additionally, HIV-1 encoded proteins influence the apoptosis in the host cell favoring or blocking T-cell apoptosis. Thus, T-cell signaling hijacked by viral proteins accounts for both viral persistence and immune suppression during HIV-1 infection. Here, we summarize past and present studies on HIV-1 T-cell signaling with special focus on the possible role of T cells in facilitating viral infection and pathogenesis
Collapse
Affiliation(s)
- Wasim Abbas
- Department of Virology, Pathogens & Inflammation Laboratory, UPRES EA4266, SFR FED 4234, University of Franche-Comte, CHRU Besançon, F-25030 Besançon, France
| | | |
Collapse
|
28
|
Klatt NR, Silvestri G, Hirsch V. Nonpathogenic simian immunodeficiency virus infections. Cold Spring Harb Perspect Med 2013; 2:a007153. [PMID: 22315718 DOI: 10.1101/cshperspect.a007153] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The simian immunodeficiency viruses (SIVs) are a diverse group of viruses that naturally infect a wide range of African primates, including African green monkeys (AGMs) and sooty mangabey monkeys (SMs). Although natural infection is widespread in feral populations of AGMs and SMs, this infection generally does not result in immunodeficiency. However, experimental inoculation of Asian macaques results in an immunodeficiency syndrome remarkably similar to human AIDS. Thus, natural nonprogressive SIV infections appear to represent an evolutionary adaptation between these animals and their primate lentiviruses. Curiously, these animals maintain robust virus replication but have evolved strategies to avoid disease progression. Adaptations observed in these primates include phenotypic changes to CD4(+) T cells, limited chronic immune activation, and altered mucosal immunity. It is probable that these animals have achieved a unique balance between T-cell renewal and proliferation and loss through activation-induced apoptosis, and virus-induced cell death. A clearer understanding of the mechanisms underlying the lack of disease progression in natural hosts for SIV infection should therefore yield insights into the pathogenesis of AIDS and may inform vaccine design.
Collapse
Affiliation(s)
- Nichole R Klatt
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
29
|
Divergent kinetics of proliferating T cell subsets in simian immunodeficiency virus (SIV) infection: SIV eliminates the "first responder" CD4+ T cells in primary infection. J Virol 2013; 87:7032-8. [PMID: 23596288 DOI: 10.1128/jvi.00027-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although increased lymphocyte turnover in chronic human immunodeficiency virus and simian immunodeficiency virus (SIV) infection has been reported in blood, there is little information on cell turnover in tissues, particularly in primary SIV infection. Here we examined the levels of proliferating T cell subsets in mucosal and peripheral lymphoid tissues of adult macaques throughout SIV infection. To specifically label cells in S-phase division, all animals were inoculated with bromodeoxyuridine 24 h prior to sampling. In healthy macaques, the highest levels of proliferating CD4(+) and CD8(+) T cells were in blood and, to a lesser extent, in spleen. Substantial percentages of proliferating cells were also found in intestinal tissues, including the jejunum, ileum, and colon, but very few proliferating cells were detected in lymph nodes (axillary and mesenteric). Moreover, essentially all proliferating T cells in uninfected animals coexpressed CD95 and many coexpressed CCR5 in the tissues examined. Confocal microscopy also demonstrated that proliferating cells were substantial viral target cells for SIV infection and viral replication. After acute SIV infection, percentages of proliferating CD4(+) and CD8(+) T cells were significantly higher in tissues of chronically infected macaques and macaques with AIDS than in those of the controls. Surprisingly, however, we found that proliferating CD4(+) T cells were selectively decreased in very early infection (8 to 10 days postinoculation [dpi]). In contrast, levels of proliferating CD8(+) T cells rapidly increased after SIV infection, peaked by 13 to 21 dpi, and thereafter remained significantly higher than those in the controls. Taken together, these findings suggest that SIV selectively infects and destroys dividing, nonspecific CD4(+) T cells in acute infection, resulting in homeostatic changes and perhaps continuing loss of replication capacity to respond to nonspecific and, later, SIV-specific antigens.
Collapse
|
30
|
Walker TN, Cimakasky LM, Coleman EM, Madison MN, Hildreth JE. Antibody against integrin lymphocyte function-associated antigen 1 inhibits HIV type 1 infection in primary cells through caspase-8-mediated apoptosis. AIDS Res Hum Retroviruses 2013; 29:371-83. [PMID: 22697794 DOI: 10.1089/aid.2011.0395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
HIV-1 infection induces formation of a virological synapse wherein CD4, chemokine receptors, and cell-adhesion molecules such as lymphocyte function-associated antigen 1 (LFA-1) form localized domains on the cell surface. Studies show that LFA-1 on the surface of HIV-1 particles retains its adhesion function and enhances virus attachment to susceptible cells by binding its counterreceptor intercellular adhesion molecule 1 (ICAM-1). This virus-cell interaction augments virus infectivity by facilitating binding and entry events. In this study, we demonstrate that inhibition of the LFA-1/ICAM-1 interaction by a monoclonal antibody leads to decreased virus production and spread in association with increased apoptosis of HIV-infected primary T cells. The data indicate that the LFA-1/ICAM-1 interaction may limit apoptosis in HIV-1-infected T cells. This phenomenon appears similar to anoikis wherein epithelial cells are protected from apoptosis conferred by ligand-bound integrins. These results have implications for further understanding HIV pathogenesis and replication in peripheral compartments and lymphoid organs.
Collapse
Affiliation(s)
- Tiffany N. Walker
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee
| | | | - Ebony M. Coleman
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, California
| | - M. Nia Madison
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee
| | - James E.K. Hildreth
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, California
| |
Collapse
|
31
|
Distinct gene-expression profiles associated with the susceptibility of pathogen-specific CD4 T cells to HIV-1 infection. Blood 2012; 121:1136-44. [PMID: 23258923 DOI: 10.1182/blood-2012-07-446278] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In HIV infection, CD4 responses to opportunistic pathogens such as Candida albicans are lost early, but CMV-specific CD4 response persists. Little is currently known about HIV infection of CD4 T cells of different pathogen/antigen specificities. CFSE-labeled PBMCs were stimulated with CMV, tetanus toxoid (TT), and C albicans antigens and subsequently exposed to HIV. HIV infection was monitored by intracellular p24 in CFSE(low) population. We found that although TT- and C albicans-specific CD4 T cells were permissive, CMV-specific CD4 T cells were highly resistant to both R5 and X4 HIV. Quantification of HIV DNA in CFSE(low) cells showed a reduction of strong-stop and full-length DNA in CMV-specific cells compared with TT- and C albicans-specific cells. β-Chemokine neutralization enhanced HIV infection in TT- and C albicans-specific cells, whereas HIV infection in CMV-specific cells remained low despite increased entry by β-chemokine neutralization, suggesting postentry HIV restriction by CMV-specific cells. Microarray analysis (Gene Expression Omnibus accession number: GSE42853) revealed distinct transcriptional profiles that involved selective up-regulation of comprehensive innate antiviral genes in CMV-specific cells, whereas TT- and C albicans-specific cells mainly up-regulated Th17 inflammatory response. Our data suggest a mechanism for the persistence of CMV-specific CD4 response and earlier loss of mucosal Th17-associated TT- and C albicans-specific CD4 response in AIDS.
Collapse
|
32
|
Immunogenicity of a recombinant measles-HIV-1 clade B candidate vaccine. PLoS One 2012; 7:e50397. [PMID: 23226275 PMCID: PMC3511521 DOI: 10.1371/journal.pone.0050397] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/19/2012] [Indexed: 01/08/2023] Open
Abstract
Live attenuated measles virus is one of the most efficient and safest vaccines available, making it an attractive candidate vector for a HIV/AIDS vaccine aimed at eliciting cell-mediated immune responses (CMI). Here we have characterized the potency of CMI responses generated in mice and non-human primates after intramuscular immunisation with a candidate recombinant measles vaccine carrying an HIV-1 insert encoding Clade B Gag, RT and Nef (MV1-F4). Eight Mauritian derived, MHC-typed cynomolgus macaques were immunised with 105 TCID50 of MV1-F4, four of which were boosted 28 days later with the same vaccine. F4 and measles virus (MV)-specific cytokine producing T cell responses were detected in 6 and 7 out of 8 vaccinees, respectively. Vaccinees with either M6 or recombinant MHC haplotypes demonstrated the strongest cytokine responses to F4 peptides. Polyfunctional analysis revealed a pattern of TNFα and IL-2 responses by CD4+ T cells and TNFα and IFNγ responses by CD8+ T cells to F4 peptides. HIV-specific CD4+ and CD8+ T cells expressing cytokines waned in peripheral blood lymphocytes by day 84, but CD8+ T cell responses to F4 peptides could still be detected in lymphoid tissues more than 3 months after vaccination. Anti-F4 and anti-MV antibody responses were detected in 6 and 8 out of 8 vaccinees, respectively. Titres of anti-F4 and MV antibodies were boosted in vaccinees that received a second immunisation. MV1-F4 carrying HIV-1 Clade B inserts induces robust boostable immunity in non-human primates. These results support further exploration of the MV1-F4 vector modality in vaccination strategies that may limit HIV-1 infectivity.
Collapse
|
33
|
Malzahn J, Shen C, Caruso L, Ghosh P, Sankapal SR, Barratt-Boyes S, Gupta P, Chen Y. Effect of early anti-retroviral therapy on the pathogenic changes in mucosal tissues of SIV infected rhesus macaques. Virol J 2012; 9:269. [PMID: 23150992 PMCID: PMC3570381 DOI: 10.1186/1743-422x-9-269] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 11/12/2012] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The gastrointestinal tissue plays an important role in the pathogenesis of HIV/SIV infection and serves as a viral reservoir in infected individuals under antiretroviral therapy (ART). However, the effect of ART administration in the very early stage of infection on HIV/SIV replication and pathogenesis in gastrointestinal tissue has not been fully studied. In this current study, rhesus monkeys infected with SIV were treated with ART starting at day 7 post-infection. The effect of early ART on SIV replication and infection-related pathogenic changes in mucosal tissues of the infected monkeys was examined. METHODS Nuclear acids were extracted from snap frozen ileum and colon tissues and mesentery lymph nodes from SIV infected monkeys with or without ART. SIV RNA and DNA loads as well as levels of CD3, CD4 and cytokine mRNA were measured by PCR and RT PCR from the isolated nuclear acids. Tissue sections were stained by immuno-fluorescence labeled antibodies for CD3 and CD4. RESULTS Without ART treatment, these monkeys underwent a mild SIV infection with low viral loads and slightly decreased CD4+ T cell counts in peripheral blood. In ART treated monkeys, SIV RNA loads were undetectable in blood with normal CD4+ T cell counts, however, SIV RNA and DNA were detected in the intestinal tissues and mesentery lymph nodes although the levels were lower than those in untreated monkeys. The levels of CD3 and CD4 positive cells in the tissues were similar between the infected untreated monkeys and infected ART treated monkeys based on RT-PCR and immune-fluorescence staining of the tissue sections. Furthermore, compatible levels of IL-6, TNF-a, IL-1b and MyD88 mRNAs were detected in most of intestinal tissues and mesentery lymph nodes of infected ART treated and infected untreated monkeys. CONCLUSIONS These results suggest that early ART administration could not effectively inhibit SIV replication in intestinal tissues and mesentery lymph nodes and could not reduce the immune activation induced by SIV infection in the intestinal tissues.
Collapse
Affiliation(s)
- Jessica Malzahn
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Inhibitory effect of HIV-specific neutralizing IgA on mucosal transmission of HIV in humanized mice. Blood 2012; 120:4571-82. [PMID: 23065154 DOI: 10.1182/blood-2012-04-422303] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
HIV-1 infections are generally initiated at mucosal sites. Thus, IgA antibody, which plays pivotal roles in mucosal immunity, might efficiently prevent HIV infection. However, mounting a highly effective HIV-specific mucosal IgA response by conventional immunization has been challenging and the potency of HIV-specific IgA against infection needs to be addressed in vivo. Here we show that the polymeric IgA form of anti-HIV antibody inhibits HIV mucosal transmission more effectively than the monomeric IgA or IgG1 form in a comparable range of concentrations in humanized mice. To deliver anti-HIV IgA in a continual manner, we devised a hematopoietic stem/progenitor cell (HSPC)-based genetic approach using an IgA gene. We transplanted human HSPCs transduced with a lentiviral construct encoding a class-switched anti-HIV IgA (b12-IgA) into the humanized bone marrow-liver-thymus (BLT) mice. The transgene was expressed specifically in B cells and plasma cells in lymphoid organs and mucosal sites. After vaginal HIV-1 challenge, mucosal CD4(+) T cells in the b12-IgA-producing mice were protected from virus-mediated depletion. Similar results were also obtained in a second humanized model, "human immune system mice." Our study demonstrates the potential of anti-HIV IgA in immunoprophylaxis in vivo, emphasizing the importance of the mucosal IgA response in defense against HIV/AIDS.
Collapse
|
35
|
Stebbings R, Eastwood D, Poole S, Thorpe R. After TGN1412: recent developments in cytokine release assays. J Immunotoxicol 2012; 10:75-82. [PMID: 22967038 PMCID: PMC3541671 DOI: 10.3109/1547691x.2012.711783] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The failure of regulatory science to keep pace with and support the development of new biological medicines was very publically highlighted in March 2006 when the first-in-man Phase I clinical trial of the immunomodulatory CD28-specific monoclonal antibody (mAb) TGN1412 ended in disaster when all six volunteers suffered a life-threatening adverse reaction termed a 'Cytokine Storm'. The poor predictive value of standard pre-clinical safety tests and animal models applied to TGN1412 demonstrated the need for a new generation of immunotoxicity assays and animal models that are both sensitive and predictive of clinical outcome in man. The non-predictive result obtained from pre-clinical safety testing in cynomolgus macaques has now been attributed to a lack of CD28 expression on CD4+ effector memory T-cells that therefore cannot be stimulated by TGN1412. In contrast, high levels of CD28 are expressed on human CD4+ effector memory T-cells, the source of most TGN1412-stimulated pro-inflammatory cytokines. Standard in vitro safety tests with human cells were also non-predictive as they did not replicate in vivo presentation of TGN1412. It was subsequently shown that, if an immobilized therapeutic mAb-based assay or endothelial cell co-culture assay was used to evaluate TGN1412, then these would have predicted a pro-inflammatory response in man. New in vitro assays based on these approaches are now being applied to emerging therapeutics to hopefully prevent a repeat of the TGN1412 incident. It has emerged that the mechanism of pro-inflammatory cytokine release stimulated by TGN1412 is different to that of other therapeutic mAbs, such that standard pro-inflammatory markers such as TNFα and IL-8 are not discriminatory. Rather, IL-2 release and lymphoproliferation are optimal readouts of a TGN1412-like pro-inflammatory response.
Collapse
Affiliation(s)
- R Stebbings
- Biotherapeutics Group, NIBSC, Potters Bar, Hertfordshire EN6 3QG, UK.
| | | | | | | |
Collapse
|
36
|
Interaction between HIV and Mycobacterium tuberculosis: HIV-1-induced CD4 T-cell depletion and the development of active tuberculosis. Curr Opin HIV AIDS 2012; 7:268-75. [PMID: 22495739 DOI: 10.1097/coh.0b013e3283524e32] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW HIV infection is the main driver of the HIV/tuberculosis (TB) syndemic in southern Africa since the early 1990s, when HIV infection rates started to increase exponentially and TB incidence rates quadruplet simultaneously. Here, we discuss pathogenic mechanisms of HIV-induced CD4 T-cell depletion and their potential impact on immune control of Mycobacterium tuberculosis. RECENT FINDINGS Depletion of effector memory CD4 T cells from the air-tissue interphase, their dysfunctional regeneration and the preferential depletion of MTB-specific CD4 T cells from circulation and from the air-tissue interphase might be key factors for the increased susceptibility to develop active TB after HIV infection. SUMMARY Early initiation of antiretroviral therapy or the development of an efficacious HIV vaccine would be the best options to reduce morbidity and mortality associated with the HIV/TB syndemic.
Collapse
|
37
|
Abstract
The complex interplay between the host immune response and HIV has been the subject of intense research over the last 25 years. HIV and simian immunodeficiency virus (SIV) CD8 T cells have been of particular interest since they were demonstrated to be temporally associated with reduction in virus load shortly following transmission. Here, we briefly review the phenotypic and functional properties of HIV-specific and SIV-specific CD8 T-cell subsets during HIV infection and consider the influence of viral variation with specific responses that are associated with disease progression or control. The development of an effective HIV/AIDS vaccine combined with existing successful prevention and treatment strategies is essential for preventing new infections. In the context of previous clinical HIV/AIDS vaccine trials, we consider the challenges faced by therapeutic and vaccine strategies designed to elicit effective HIV-specific CD8 T cells.
Collapse
|
38
|
Tenaya IWM, Heel K, Stumbles PA, Wilcox GE. Flow cytometric analysis of lymphocyte subset kinetics in Bali cattle experimentally infected with Jembrana disease virus. Vet Immunol Immunopathol 2012; 149:167-76. [PMID: 22776774 DOI: 10.1016/j.vetimm.2012.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 04/24/2012] [Accepted: 06/11/2012] [Indexed: 11/17/2022]
Abstract
Jembrana disease virus (JDV) is an unusual bovine lentivirus that causes an acute and sometimes fatal disease after a short incubation period in Bali cattle (Bos javanicus). The pathological changes occur primarily in lymphoid tissues, which feature proliferating lymphoblastoid-like cells predominantly throughout parafollicular (T-cell) areas, and atrophy of follicles (B-cell) areas. Five Bali cattle were experimentally infected with JDV and all developed typical clinical signs of Jembrana disease characterised by a transient febrile response, enlargement of superficial lymph nodes and a significant leukopenia. Flow cytometric analysis of PBMC during the acute (febrile) disease phase showed that the reduced number of lymphocytes was due to a significant decrease in both the proportion and absolute numbers of CD4(+) T cells, but not CD8(+) T-cells or CD21(+) B-cells. At the end of the febrile phase, total numbers of both CD8(+) T-cells and CD21(+) B-cells increased significantly, while CD4(+) T-cell numbers remained below normal values, resulting in a significantly reduced CD4(+):CD8(+) ratio. We speculate that the persistent depletion of CD4(+) T cells following JDV infection, through lack of CD4(+) T cell help to B cells, may explain the lack of production of JDV-specific antibodies for several weeks after recovery despite an increase in CD21(+) B cell numbers. Further, our previous data showing that IgG(+) plasma cells are targets for JDV infection, correlated with our current data demonstrating an increase in CD8(+) T cell numbers, supports the suggestion that anti-viral cytotoxic T cell or other cell-mediated immune responses may be critical in the recovery process, although this remains to be formally demonstrated for JDV.
Collapse
Affiliation(s)
- I W Masa Tenaya
- School of Veterinary and Biomedical Sciences, Murdoch University, Murdoch, WA 6150, Australia
| | | | | | | |
Collapse
|
39
|
Kinetic model of HIV infection including hematopoietic progenitor cells. Math Biosci 2012; 236:36-43. [DOI: 10.1016/j.mbs.2012.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 01/10/2012] [Accepted: 01/13/2012] [Indexed: 12/16/2022]
|
40
|
Interferon-alpha administration enhances CD8+ T cell activation in HIV infection. PLoS One 2012; 7:e30306. [PMID: 22291932 PMCID: PMC3265460 DOI: 10.1371/journal.pone.0030306] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 12/13/2011] [Indexed: 12/11/2022] Open
Abstract
Background Type I interferons play important roles in innate immune defense. In HIV infection, type I interferons may delay disease progression by inhibiting viral replication while at the same time accelerating disease progression by contributing to chronic immune activation. Methods To investigate the effects of type I interferons in HIV-infection, we obtained cryopreserved peripheral blood mononuclear cell samples from 10 subjects who participated in AIDS Clinical Trials Group Study 5192, a trial investigating the activity of systemic administration of IFNα for twelve weeks to patients with untreated HIV infection. Using flow cytometry, we examined changes in cell cycle status and expression of activation antigens by circulating T cells and their maturation subsets before, during and after IFNα treatment. Results The proportion of CD38+HLA-DR+CD8+ T cells increased from a mean of 11.7% at baseline to 24.1% after twelve weeks of interferon treatment (p = 0.006). These frequencies dropped to an average of 20.1% six weeks after the end of treatment. In contrast to CD8+ T cells, the frequencies of activated CD4+ T cells did not change with administration of type I interferon (mean percentage of CD38+DR+ cells = 2.62% at baseline and 2.17% after 12 weeks of interferon therapy). As plasma HIV levels fell with interferon therapy, this was correlated with a “paradoxical” increase in CD8+ T cell activation (p<0.001). Conclusion Administration of type I interferon increased expression of the activation markers CD38 and HLA DR on CD8+ T cells but not on CD4+ T cells of HIV+ persons. These observations suggest that type I interferons may contribute to the high levels of CD8+ T cell activation that occur during HIV infection.
Collapse
|
41
|
Boyapalle S, Mohapatra S, Mohapatra S. Nanotechnology Applications to HIV Vaccines and Microbicides. J Glob Infect Dis 2012; 4:62-8. [PMID: 22529630 PMCID: PMC3326962 DOI: 10.4103/0974-777x.93764] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) remains one of the most serious threats to global health. Today there are no HIV vaccines which can prevent HIV infection. All of the candidates being studied are in the experimental stage. Preventive vaccine candidates are being tested in HIV-negative people to see if they can prevent infection. With of the development of a safe and effective vaccine still likely to be years away, topical microbicide formulations that are applied vaginally and rectally are receiving greater interest as an effective alternative to slow down the global spread of HIV. Current microbicide trials that aim to prevent the sexual transmission of HIV are using gels, creams, rings, films and there is also work underway to explore other types of 'delivery' systems. There have been numerous reports on safety and lack of toxicity of the application of nanotechnology for targeted delivery and slow, sustained release of drugs, proteins, peptides or nucleic acids by any route to maximize effectiveness and minimize adverse effects. The application of nanotechnology for targeting drugs and macromolecules to specific tissues or cells is one of the most important areas in nanomedicine research. Thus far nanoparticles provide a strong platform to combine protein and DNA based vaccines/microbicides and will facilitate the production, preclinical evaluation and clinical testing in the future.
Collapse
Affiliation(s)
- Sandhya Boyapalle
- Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Subhra Mohapatra
- Nanomedicine Research Center, University of South Florida Morsani College of Medicine, Tampa, FL
- Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Shyam Mohapatra
- Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL
- Nanomedicine Research Center, University of South Florida Morsani College of Medicine, Tampa, FL
- James A Haley Veteran's Administration Hospital and Medical Center, VA Hospital, Tampa, FL
| |
Collapse
|
42
|
Taaffe JE, Bosinger SE, Del Prete GQ, Else JG, Ratcliffe S, Ward CD, Migone T, Paiardini M, Silvestri G. CCR5 blockade is well tolerated and induces changes in the tissue distribution of CCR5+ and CD25+ T cells in healthy, SIV-uninfected rhesus macaques. J Med Primatol 2011; 41:24-42. [PMID: 22077380 DOI: 10.1111/j.1600-0684.2011.00521.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND CCR5 is a main co-receptor for HIV, but also homes lymphocytes to sites of inflammation. We hypothesized that inhibition of CCR5 signaling would reduce HIV-associated chronic immune activation. METHODS To test this hypothesis, we administered an antagonistic anti-CCR5 monoclonal antibody (HGS101) to five uninfected rhesus macaques (RMs) and monitored lymphocyte dynamics in blood and tissue. RESULTS CCR5 blockade resulted in decreased levels of CCR5+ T cells in blood and, at later timepoints, in lymph nodes. Additionally, the levels of CD25+ T cells increased in lymph nodes, but decreased in blood, bone marrow, and rectal mucosa. Finally, a profile of gene expression from HGS101-treated RMs revealed a subtle, but consistent, in vivo signature of CCR5 blockade that suggests a mild immune-modulatory effect. CONCLUSIONS Treatment with anti-CCR5 antibody induces changes in the tissue distribution of CCR5+ and CD25+ T cells that may impact on the overall levels of immune activation during HIV and SIV infection.
Collapse
Affiliation(s)
- Jessica E Taaffe
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Distinct expression patterns of CD69 in mucosal and systemic lymphoid tissues in primary SIV infection of rhesus macaques. PLoS One 2011; 6:e27207. [PMID: 22096538 PMCID: PMC3212564 DOI: 10.1371/journal.pone.0027207] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 10/11/2011] [Indexed: 12/04/2022] Open
Abstract
Although the intestinal tract plays a major role in early human immunodeficiency virus (HIV) infection, the role of immune activation and viral replication in intestinal tissues is not completely understood. Further, increasing evidence suggests the early leukocyte activation antigen CD69 may be involved in the development or regulation of important T cell subsets, as well as a major regulatory molecule of immune responses. Using the simian immunodeficiency virus (SIV) rhesus macaque model, we compared expression of CD69 on T cells from the intestine, spleen, lymph nodes, and blood of normal and SIV-infected macaques throughout infection. In uninfected macaques, the majority of intestinal lamina propria CD4+ T cells had a memory (CD95+) phenotype and co-expressed CD69, and essentially all intestinal CCR5+ cells co-expressed CD69. In contrast, systemic lymphoid tissues had far fewer CD69+ T cells, and many had a naïve phenotype. Further, marked, selective depletion of intestinal CD4+CD69+ T cells occurred in early SIV infection, and this depletion persisted throughout infection. Markedly increased levels of CD8+CD69+ T cells were detected after SIV infection in virtually all tissues, including the intestine. Further, confocal microscopy demonstrated selective, productive infection of CD3+CD69+ T cells in the intestine in early infection. Combined, these results indicate CD69+CD4+ T cells are a major early target for viral infection, and their rapid loss by direct infection may have profound effects on intestinal immune regulation in HIV infected patients.
Collapse
|
44
|
Ortiz AM, Klatt NR, Li B, Yi Y, Tabb B, Hao XP, Sternberg L, Lawson B, Carnathan PM, Cramer EM, Engram JC, Little DM, Ryzhova E, Gonzalez-Scarano F, Paiardini M, Ansari AA, Ratcliffe S, Else JG, Brenchley JM, Collman RG, Estes JD, Derdeyn CA, Silvestri G. Depletion of CD4⁺ T cells abrogates post-peak decline of viremia in SIV-infected rhesus macaques. J Clin Invest 2011; 121:4433-45. [PMID: 22005304 PMCID: PMC3204830 DOI: 10.1172/jci46023] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 09/07/2011] [Indexed: 12/13/2022] Open
Abstract
CD4+ T cells play a central role in the immunopathogenesis of HIV/AIDS, and their depletion during chronic HIV infection is a hallmark of disease progression. However, the relative contribution of CD4+ T cells as mediators of antiviral immune responses and targets for virus replication is still unclear. Here, we have generated data in SIV-infected rhesus macaques (RMs) that suggest that CD4+ T cells are essential in establishing control of virus replication during acute infection. To directly assess the role of CD4+ T cells during primary SIV infection, we in vivo depleted these cells from RMs prior to infecting the primates with a pathogenic strain of SIV. Compared with undepleted animals, CD4+ lymphocyte-depleted RMs showed a similar peak of viremia, but did not manifest any post-peak decline of virus replication despite CD8+ T cell- and B cell-mediated SIV-specific immune responses comparable to those observed in control animals. Interestingly, depleted animals displayed rapid disease progression, which was associated with increased virus replication in non-T cells as well as the emergence of CD4-independent SIV-envelopes. Our results suggest that the antiviral CD4+ T cell response may play an important role in limiting SIV replication, which has implications for the design of HIV vaccines.
Collapse
Affiliation(s)
- Alexandra M. Ortiz
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nichole R. Klatt
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Bing Li
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yanjie Yi
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Brian Tabb
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Xing Pei Hao
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lawrence Sternberg
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Benton Lawson
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paul M. Carnathan
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elizabeth M. Cramer
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jessica C. Engram
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dawn M. Little
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elena Ryzhova
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Francisco Gonzalez-Scarano
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mirko Paiardini
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Aftab A. Ansari
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sarah Ratcliffe
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - James G. Else
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jason M. Brenchley
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ronald G. Collman
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jacob D. Estes
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cynthia A. Derdeyn
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
45
|
Spudich S, Gisslen M, Hagberg L, Lee E, Liegler T, Brew B, Fuchs D, Tambussi G, Cinque P, Hecht FM, Price RW. Central nervous system immune activation characterizes primary human immunodeficiency virus 1 infection even in participants with minimal cerebrospinal fluid viral burden. J Infect Dis 2011; 204:753-60. [PMID: 21844301 DOI: 10.1093/infdis/jir387] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Central nervous system (CNS) human immunodeficiency virus (HIV) infection and immune activation lead to brain injury and neurological impairment. Although HIV enters the nervous system soon after transmission, the magnitude of infection and immunoactivation within the CNS during primary HIV infection (PHI) has not been characterized. METHODS This cross-sectional study analyzed cerebrospinal fluid (CSF) and blood from 96 participants with PHI and compared them with samples from neuroasymptomatic participants with chronic infection and ≥ 200 or < 200 blood CD4 T cells/μL, and with samples from HIV-seronegative participants with respect to CSF and plasma HIV RNA, CSF to serum albumin ratio, and CSF white blood cell counts (WBC), neopterin levels, and concentrations of chemokines CXCL10 and CCL2. RESULTS The PHI participants (median 77 days post transmission) had CSF HIV RNA, WBC, neopterin, and CXCL10 concentrations similar to the chronic infection participants but uniquely high albumin ratios. 18 participants had ≤ 100 copies/mL CSF HIV RNA, which was associated with low CSF to plasma HIV ratios and levels of CSF inflammation lower than in other PHI participants but higher than in HIV-seronegative controls. CONCLUSIONS Prominent CNS infection and immune activation is evident during the first months after HIV transmission, though a proportion of PHI patients demonstrate relatively reduced CSF HIV RNA and inflammation during this early period.
Collapse
Affiliation(s)
- Serena Spudich
- Department of Neurology, University of California San Francisco, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
CD4 T cell depletion at the cervix during HIV infection is associated with accumulation of terminally differentiated T cells. J Virol 2011; 85:13333-41. [PMID: 21994461 DOI: 10.1128/jvi.05671-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In blood, the accumulation of terminally differentiated (TD) T cells during HIV infection is associated with CD4 T cell loss and HIV disease progression. Here, we investigated the maintenance and functional characteristics of memory T cells at the cervix. We found that CD4 T cell depletion at the cervix mirrors CD4 depletion in blood. In all women, depletion of CD4 T cells at the cervix was associated with significant reductions in CD45RA- CCR7+ (central memory [CM]) T cells and the accumulation of CD45RA+ CCR7- (TD T cells). We determined whether inflammation in the genital tract was associated with the local differentiation of T cells at the cervix. In uninfected women, genital tract inflammation was associated with the accumulation of CD45RA- CCR7+ CM CD4 T cells and reduced frequencies of CD45RA+ CCR7- TD cells at the cervix. This finding may reflect the fact that, in the absence of HIV infection, TD T cells may be slowly lost in the presence of genital inflammation, while CD45RA- CCR7+ CM T cells are recruited to replenish the diminishing CD4 T cell pool. Following global stimulation with phorbol myristate acetate (PMA)-ionomycin, we noted a significant interleukin 2 (IL-2) deficit in both cervical and blood CD4 T cells from HIV-infected women compared to uninfected women, while gamma interferon (IFN-γ) production was similar, irrespective of HIV status. Few HIV-infected women had detectable IFN-γ and IL-2 HIV-specific T cell responses at the cervix, and these responses were significantly lower in magnitude than the corresponding responses in blood. These data suggest that CD4 depletion was associated with the accumulation of terminally differentiated T cell phenotypes at the cervical mucosa defective in their ability to produce IL-2. CD4 depletion and compromised immunity at the cervix may be accompanied by progressive decline of central memory-like T cells and development of T cells toward terminally differentiated phenotypes.
Collapse
|
47
|
Sensitivity to electrical stimulation of human immunodeficiency virus type 1 and MAGIC-5 cells. AMB Express 2011; 1:23. [PMID: 21906386 PMCID: PMC3222307 DOI: 10.1186/2191-0855-1-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 08/08/2011] [Indexed: 11/15/2022] Open
Abstract
To determine the sensitivities to low electrical potential of human immunodeficiency virus type 1 (HIV-1) and its target cells, HIV-1 and MAGIC-5 cells were directly stimulated with a constant direct current potential of 1.0 V (vs. Ag/AgCl). HIV-1 was incubated for 3 h at 37°C on a poly-L-lysine-coated indium-tin oxide electrode, and then stimulated by an electrical potential. MAGIC-5 cells were seeded onto the electrically stimulated HIV-1 and cultured for 3 days at 37°C. HIV-1-infected cells were measured by multinuclear activation via a galactosidase indicator assay. MAGIC-5 cells were also stimulated by an electrical potential of 1.0 V; cell damage, proliferation and apoptosis were evaluated by trypan blue staining, cell counting and in situ apoptosis detection, respectively. HIV-1 was found to be damaged to a greater extent by electrical stimulation than the cells. In particular, after application of a 1.0-V potential for 3 min, HIV-1LAI and HIV-1KMT infection were inhibited by about 90%, but changes in cell damage, proliferation and apoptosis were virtually undetectable. These results suggested that HIV-1 is significantly more susceptible to low electrical potential than cells. This finding could form the basis of a novel therapeutic strategy against HIV-1 infection.
Collapse
|
48
|
Nowak RG, Gravitt PE, Morrison CS, Gange SJ, Kwok C, Oliver AE, Howard R, Van der Pol B, Salata RA, Padian NS, Chipato T, Munjoma M, Celentano DD. Increases in human papillomavirus detection during early HIV infection among women in Zimbabwe. J Infect Dis 2011; 203:1182-91. [PMID: 21451006 DOI: 10.1093/infdis/jiq172] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Individuals who acquire human immunodeficiency virus (HIV) may experience an immediate disruption of genital tract immunity, altering the ability to mount a local and effective immune response. This study examined the impact of early HIV infection on new detection of human papillomavirus (HPV). METHODS One hundred fifty-five Zimbabwean women with observation periods before and after HIV acquisition and 486 HIV-uninfected women were selected from a cohort study evaluating hormonal contraceptive use and risk of HIV acquisition. Study visits occurred at 3-month intervals. Cervical swab samples available from up to 6 months before, at, and up to 6 months after the visit when HIV was first detected were typed for 37 HPV genotypes or subtypes. RESULTS We observed ∼5-fold higher odds of multiple (≥2) new HPV detections only after HIV acquisition, relative to HIV-negative women after adjusting for sexual behavior and concurrent genital tract infections. We also observed ∼2.5-fold higher odds of single new HPV detections at visits before and after HIV acquisition, relative to HIV-uninfected women in multivariable models. CONCLUSIONS These findings suggest that HIV infection has an immediate impact on genital tract immunity, as evidenced by the high risk of multiple new HPV detections immediately after HIV acquisition.
Collapse
Affiliation(s)
- Rebecca G Nowak
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Martinelli E, Tharinger H, Frank I, Arthos J, Piatak M, Lifson JD, Blanchard J, Gettie A, Robbiani M. HSV-2 infection of dendritic cells amplifies a highly susceptible HIV-1 cell target. PLoS Pathog 2011; 7:e1002109. [PMID: 21738472 PMCID: PMC3128120 DOI: 10.1371/journal.ppat.1002109] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 04/23/2011] [Indexed: 11/18/2022] Open
Abstract
Herpes simplex virus type 2 (HSV-2) increases the risk of HIV-1 infection and, although several reports describe the interaction between these two viruses, the exact mechanism for this increased susceptibility remains unclear. Dendritic cells (DCs) at the site of entry of HSV-2 and HIV-1 contribute to viral spread in the mucosa. Specialized DCs present in the gut-associated lymphoid tissues produce retinoic acid (RA), an important immunomodulator, able to influence HIV-1 replication and a key mediator of integrin α₄β₇ on lymphocytes. α₄β₇ can be engaged by HIV-1 on the cell-surface and CD4⁺ T cells expressing high levels of this integrin (α₄β₇ (high)) are particularly susceptible to HIV-1 infection. Herein we provide in-vivo data in macaques showing an increased percentage of α₄β₇ (high) CD4⁺ T cells in rectal mucosa, iliac lymph nodes and blood within 6 days of rectal exposure to live (n = 11), but not UV-treated (n = 8), HSV-2. We found that CD11c⁺ DCs are a major target of HSV-2 infection in in-vitro exposed PBMCs. We determined that immature monocyte-derived DCs (moDCs) express aldehyde dehydrogenase ALDH1A1, an enzyme essential for RA production, which increases upon HSV-2 infection. Moreover, HSV-2-infected moDCs significantly increase α₄β₇ expression on CD4⁺ T lymphocytes and HIV-1 infection in DC-T cell mixtures in a RA-dependent manner. Thus, we propose that HSV-2 modulates its microenviroment, influencing DC function, increasing RA production capability and amplifying a α₄β₇ (high)CD4⁺ T cells. These factors may play a role in increasing the susceptibility to HIV-1.
Collapse
Affiliation(s)
- Elena Martinelli
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Hugo Tharinger
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Ines Frank
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Piatak
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., National Cancer Institute, Frederick, Maryland, Unites States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., National Cancer Institute, Frederick, Maryland, Unites States of America
| | - James Blanchard
- Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, Louisiana, United States of America
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, New York, Unites States of America
| | - Melissa Robbiani
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| |
Collapse
|
50
|
Février M, Dorgham K, Rebollo A. CD4+ T cell depletion in human immunodeficiency virus (HIV) infection: role of apoptosis. Viruses 2011; 3:586-612. [PMID: 21994747 PMCID: PMC3185763 DOI: 10.3390/v3050586] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 05/03/2011] [Accepted: 05/04/2011] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection is principally a mucosal disease and the gastrointestinal (GI) tract is the major site of HIV replication. Loss of CD4+ T cells and systemic immune hyperactivation are the hallmarks of HIV infection. The end of acute infection is associated with the emergence of specific CD4+ and CD8+ T cell responses and the establishment of a chronic phase of infection. Abnormal levels of immune activation and inflammation persist despite a low steady state level of viremia. Although the causes of persistent immune hyperactivation remain incompletely characterized, physiological alterations of gastrointestinal tract probably play a major role. Failure to restore Th17 cells in gut-associated lymphoid tissues (GALT) might impair the recovery of the gut mucosal barrier. This review discusses recent advances on understanding the contribution of CD4+ T cell depletion to HIV pathogenesis.
Collapse
Affiliation(s)
- Michèle Février
- Unité Génomique Virale et Vaccination, CNRS URA3015, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France.
| | | | | |
Collapse
|