1
|
Pampusch MS, Sevcik EN, Quinn ZE, Davey BC, Berg JM, Gorrell-Brown I, Abdelaal HM, Rakasz EG, Rendahl A, Skinner PJ. Assessment of anti-CD20 antibody pre-treatment for augmentation of CAR-T cell therapy in SIV-infected rhesus macaques. Front Immunol 2023; 14:1101446. [PMID: 36825014 PMCID: PMC9941136 DOI: 10.3389/fimmu.2023.1101446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
During chronic HIV and SIV infections, the majority of viral replication occurs within lymphoid follicles. In a pilot study, infusion of SIV-specific CD4-MBL-CAR-T cells expressing the follicular homing receptor, CXCR5, led to follicular localization of the cells and a reduction in SIV viral loads in rhesus macaques. However, the CAR-T cells failed to persist. We hypothesized that temporary disruption of follicles would create space for CAR-T cell engraftment and lead to increased abundance and persistence of CAR-T cells. In this study we treated SIV-infected rhesus macaques with CAR-T cells and preconditioned one set with anti-CD20 antibody to disrupt the follicles. We evaluated CAR-T cell abundance and persistence in four groups of SIVmac239-infected and ART-suppressed animals: untreated, CAR-T cell treated, CD20 depleted, and CD20 depleted/CAR-T cell treated. In the depletion study, anti-CD20 was infused one week prior to CAR-T infusion and cessation of ART. Anti-CD20 antibody treatment led to temporary depletion of CD20+ cells in blood and partial depletion in lymph nodes. In this dose escalation study, there was no impact of CAR-T cell infusion on SIV viral load. However, in both the depleted and non-depleted animals, CAR-T cells accumulated in and around lymphoid follicles and were Ki67+. CAR-T cells increased in number in follicles from 2 to 6 days post-treatment, with a median 15.2-fold increase in follicular CAR-T cell numbers in depleted/CAR-T treated animals compared to an 8.1-fold increase in non-depleted CAR-T treated animals. The increase in CAR T cells in depleted animals was associated with a prolonged elevation of serum IL-6 levels and a rapid loss of detectable CAR-T cells. Taken together, these data suggest that CAR-T cells likely expanded to a greater extent in depleted/CAR-T cell treated animals. Further studies are needed to elucidate mechanisms mediating the rapid loss of CAR-T cells and to evaluate strategies to improve engraftment and persistence of HIV-specific CAR-T cells. The potential for an inflammatory cytokine response appears to be enhanced with anti-CD20 antibody treatment and future studies may require CRS control strategies. These studies provide important insights into cellular immunotherapy and suggest future studies for improved outcomes.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Emily N. Sevcik
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Zoe E. Quinn
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - James M. Berg
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Ian Gorrell-Brown
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison WI, United States
| | - Aaron Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
2
|
Pampusch MS, Abdelaal HM, Cartwright EK, Molden JS, Davey BC, Sauve JD, Sevcik EN, Rendahl AK, Rakasz EG, Connick E, Berger EA, Skinner PJ. CAR/CXCR5-T cell immunotherapy is safe and potentially efficacious in promoting sustained remission of SIV infection. PLoS Pathog 2022; 18:e1009831. [PMID: 35130312 PMCID: PMC8853520 DOI: 10.1371/journal.ppat.1009831] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/17/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
During chronic human immunodeficiency virus (HIV) or simian immunodeficiency virus (SIV) infection prior to AIDS progression, the vast majority of viral replication is concentrated within B cell follicles of secondary lymphoid tissues. We investigated whether infusion of T cells expressing an SIV-specific chimeric antigen receptor (CAR) and the follicular homing receptor, CXCR5, could successfully kill viral-RNA+ cells in targeted lymphoid follicles in SIV-infected rhesus macaques. In this study, CD4 and CD8 T cells from rhesus macaques were genetically modified to express antiviral CAR and CXCR5 moieties (generating CAR/CXCR5-T cells) and autologously infused into a chronically infected animal. At 2 days post-treatment, the CAR/CXCR5-T cells were located primarily in spleen and lymph nodes both inside and outside of lymphoid follicles. Few CAR/CXCR5-T cells were detected in the ileum, rectum, and lung, and no cells were detected in the bone marrow, liver, or brain. Within follicles, CAR/CXCR5-T cells were found in direct contact with SIV-viral RNA+ cells. We next infused CAR/CXCR5-T cells into ART-suppressed SIV-infected rhesus macaques, in which the animals were released from ART at the time of infusion. These CAR/CXCR5-T cells replicated in vivo within both the extrafollicular and follicular regions of lymph nodes and accumulated within lymphoid follicles. CAR/CXR5-T cell concentrations in follicles peaked during the first week post-infusion but declined to undetectable levels after 2 to 4 weeks. Overall, CAR/CXCR5-T cell-treated animals maintained lower viral loads and follicular viral RNA levels than untreated control animals, and no outstanding adverse reactions were noted. These findings indicate that CAR/CXCR5-T cell treatment is safe and holds promise as a future treatment for the durable remission of HIV.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Emily K. Cartwright
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jhomary S. Molden
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jordan D. Sauve
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Emily N. Sevcik
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Aaron K. Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, Arizona, United States of America
| | - Edward A. Berger
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| |
Collapse
|
3
|
Onabajo OO, Mattapallil JJ. Gut Microbiome Homeostasis and the CD4 T- Follicular Helper Cell IgA Axis in Human Immunodeficiency Virus Infection. Front Immunol 2021; 12:657679. [PMID: 33815419 PMCID: PMC8017181 DOI: 10.3389/fimmu.2021.657679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Human Immunodeficiency Virus (HIV) and Simian Immunodeficiency Virus (SIV) are associated with severe perturbations in the gut mucosal environment characterized by massive viral replication and depletion of CD4 T cells leading to dysbiosis, breakdown of the epithelial barrier, microbial translocation, immune activation and disease progression. Multiple mechanisms play a role in maintaining homeostasis in the gut mucosa and protecting the integrity of the epithelial barrier. Among these are the secretory IgA (sIgA) that are produced daily in vast quantities throughout the mucosa and play a pivotal role in preventing commensal microbes from breaching the epithelial barrier. These microbe specific, high affinity IgA are produced by IgA+ plasma cells that are present within the Peyer’s Patches, mesenteric lymph nodes and the isolated lymphoid follicles that are prevalent in the lamina propria of the gastrointestinal tract (GIT). Differentiation, maturation and class switching to IgA producing plasma cells requires help from T follicular helper (Tfh) cells that are present within these lymphoid tissues. HIV replication and CD4 T cell depletion is accompanied by severe dysregulation of Tfh cell responses that compromises the generation of mucosal IgA that in turn alters barrier integrity leading to commensal bacteria readily breaching the epithelial barrier and causing mucosal pathology. Here we review the effect of HIV infection on Tfh cells and mucosal IgA responses in the GIT and the consequences these have for gut dysbiosis and mucosal immunopathogenesis.
Collapse
Affiliation(s)
- Olusegun O Onabajo
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Joseph J Mattapallil
- F. E. Hebert School of Medicine, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
4
|
Abdelaal HM, Cartwright EK, Skinner PJ. Detection of Antigen-Specific T Cells Using In Situ MHC Tetramer Staining. Int J Mol Sci 2019; 20:E5165. [PMID: 31635220 PMCID: PMC6834156 DOI: 10.3390/ijms20205165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/26/2022] Open
Abstract
The development of in situ major histocompatibility complex (MHC) tetramer (IST) staining to detect antigen (Ag)-specific T cells in tissues has radically revolutionized our knowledge of the local cellular immune response to viral and bacterial infections, cancers, and autoimmunity. IST combined with immunohistochemistry (IHC) enables determination of the location, abundance, and phenotype of T cells, as well as the characterization of Ag-specific T cells in a 3-dimensional space with respect to neighboring cells and specific tissue locations. In this review, we discuss the history of the development of IST combined with IHC. We describe various methods used for IST staining, including direct and indirect IST and IST performed on fresh, lightly fixed, frozen, and fresh then frozen tissue. We also describe current applications for IST in viral and bacterial infections, cancer, and autoimmunity. IST combined with IHC provides a valuable tool for studying and tracking the Ag-specific T cell immune response in tissues.
Collapse
Affiliation(s)
- Hadia M Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA.
- Department of Microbiology and Immunology, Zagazig University, Zagazig 44519, Egypt.
| | - Emily K Cartwright
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA.
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA.
- Microbiology Research Facility, 689 23rd Avenue SE, University of Minnesota, Twin Cities, MN 55455, USA.
| |
Collapse
|
5
|
Li S, Folkvord JM, Kovacs KJ, Wagstaff RK, Mwakalundwa G, Rendahl AK, Rakasz EG, Connick E, Skinner PJ. Low levels of SIV-specific CD8+ T cells in germinal centers characterizes acute SIV infection. PLoS Pathog 2019; 15:e1007311. [PMID: 30897187 PMCID: PMC6445460 DOI: 10.1371/journal.ppat.1007311] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 04/02/2019] [Accepted: 02/08/2019] [Indexed: 11/18/2022] Open
Abstract
CD8+ T cells play an important role in controlling of HIV and SIV infections. However, these cells are largely excluded from B cell follicles where HIV and SIV producing cells concentrate during chronic infection. It is not known, however, if antigen-specific CD8+ T cells are excluded gradually as pathogenesis progresses from early to chronic phase, or this phenomenon occurs from the beginning infection. In this study we determined that SIV-specific CD8+ T cells were largely excluded from follicles during early infection, we also found that within follicles, they were entirely absent in 60% of the germinal centers (GCs) examined. Furthermore, levels of SIV-specific CD8+ T cells in follicular but not extrafollicular areas significantly correlated inversely with levels of viral RNA+ cells. In addition, subsets of follicular SIV-specific CD8+ T cells were activated and proliferating and expressed the cytolytic protein perforin. These studies suggest that a paucity of SIV-specific CD8+ T cells in follicles and complete absence within GCs during early infection may set the stage for the establishment of persistent chronic infection.
Collapse
Affiliation(s)
- Shengbin Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Joy M. Folkvord
- Division of Infectious Diseases, University of Arizona, Tucson, Arizona, United States of America
| | - Katalin J. Kovacs
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Reece K. Wagstaff
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Gwantwa Mwakalundwa
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Aaron K. Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, Arizona, United States of America
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| |
Collapse
|
6
|
McBrien JB, Kumar NA, Silvestri G. Mechanisms of CD8 + T cell-mediated suppression of HIV/SIV replication. Eur J Immunol 2018; 48:898-914. [PMID: 29427516 DOI: 10.1002/eji.201747172] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 12/13/2022]
Abstract
In this article, we summarize the role of CD8+ T cells during natural and antiretroviral therapy (ART)-treated HIV and SIV infections, discuss the mechanisms responsible for their suppressive activity, and review the rationale for CD8+ T cell-based HIV cure strategies. Evidence suggests that CD8+ T cells are involved in the control of virus replication during HIV and SIV infections. During early HIV infection, the cytolytic activity of CD8+ T cells is responsible for control of viremia. However, it has been proposed that CD8+ T cells also use non-cytolytic mechanisms to control SIV infection. More recently, CD8+ T cells were shown to be required to fully suppress virus production in ART-treated SIV-infected macaques, suggesting that CD8+ T cells are involved in the control of virus transcription in latently infected cells that persist under ART. A better understanding of the complex antiviral activities of CD8+ T cells during HIV/SIV infection will pave the way for immune interventions aimed at harnessing these functions to target the HIV reservoir.
Collapse
Affiliation(s)
- Julia Bergild McBrien
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Nitasha A Kumar
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| |
Collapse
|
7
|
Haran KP, Hajduczki A, Pampusch MS, Mwakalundwa G, Vargas-Inchaustegui DA, Rakasz EG, Connick E, Berger EA, Skinner PJ. Simian Immunodeficiency Virus (SIV)-Specific Chimeric Antigen Receptor-T Cells Engineered to Target B Cell Follicles and Suppress SIV Replication. Front Immunol 2018; 9:492. [PMID: 29616024 PMCID: PMC5869724 DOI: 10.3389/fimmu.2018.00492] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 11/13/2022] Open
Abstract
There is a need to develop improved methods to treat and potentially cure HIV infection. During chronic HIV infection, replication is concentrated within T follicular helper cells (Tfh) located within B cell follicles, where low levels of virus-specific CTL permit ongoing viral replication. We previously showed that elevated levels of simian immunodeficiency virus (SIV)-specific CTL in B cell follicles are linked to both decreased levels of viral replication in follicles and decreased plasma viral loads. These findings provide the rationale to develop a strategy for targeting follicular viral-producing (Tfh) cells using antiviral chimeric antigen receptor (CAR) T cells co-expressing the follicular homing chemokine receptor CXCR5. We hypothesize that antiviral CAR/CXCR5-expressing T cells, when infused into an SIV-infected animal or an HIV-infected individual, will home to B cell follicles, suppress viral replication, and lead to long-term durable remission of SIV and HIV. To begin to test this hypothesis, we engineered gammaretroviral transduction vectors for co-expression of a bispecific anti-SIV CAR and rhesus macaque CXCR5. Viral suppression by CAR/CXCR5-transduced T cells was measured in vitro, and CXCR5-mediated migration was evaluated using both an in vitro transwell migration assay, as well as a novel ex vivo tissue migration assay. The functionality of the CAR/CXCR5 T cells was demonstrated through their potent suppression of SIVmac239 and SIVE660 replication in in vitro and migration to the ligand CXCL13 in vitro, and concentration in B cell follicles in tissues ex vivo. These novel antiviral immunotherapy products have the potential to provide long-term durable remission (functional cure) of HIV and SIV infections.
Collapse
Affiliation(s)
- Kumudhini Preethi Haran
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Agnes Hajduczki
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Mary S Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Gwantwa Mwakalundwa
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Diego A Vargas-Inchaustegui
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, AZ, United States
| | - Edward A Berger
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
8
|
Li S, Mwakalundwa G, Skinner PJ. In Situ MHC-tetramer Staining and Quantitative Analysis to Determine the Location, Abundance, and Phenotype of Antigen-specific CD8 T Cells in Tissues. J Vis Exp 2017. [PMID: 28994787 DOI: 10.3791/56130] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
T cells are critical to many immunological processes, including detecting and eliminating virus-infected cells, preventing autoimmunity, assisting in B-cell and plasma-cell production of antibodies, and detecting and eliminating cancer cells. The development of MHC-tetramer staining of antigen-specific T cells analyzed by flow cytometry has revolutionized our ability to study and understand the immunobiology of T cells. While extremely useful for determining the quantity and phenotype of antigen-specific T cells, flow cytometry cannot determine the spatial localization of antigen-specific T cells to other cells and structures in tissues, and current disaggregation techniques to extract the T cells needed for flow cytometry have limited effectiveness in non-lymphoid tissues. In situ MHC-tetramer staining (IST) is a technique to visualize T cells that are specific for antigens of interest in tissues. In combination with immunohistochemistry (IHC), IST can determine the abundance, location, and phenotype of antigen-specific CD8 and CD4 T cells in tissues. Here, we describe a protocol to stain and enumerate antigen-specific CD8 T cells, with specific phenotypes located within specific tissue compartments. These procedures are the same that we used in our recent publication by Li et al., entitled "Simian Immunodeficiency Virus-Producing Cells in Follicles Are Partially Suppressed by CD8+ Cells In Vivo." The methods described are broadly applicable because they can be used to localize, phenotype, and quantify essentially any antigen-specific CD8 T cell for which MHC tetramers are available, in any tissue.
Collapse
Affiliation(s)
- Shengbin Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota
| | | | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota;
| |
Collapse
|
9
|
Distinct transcriptome profiles of Gag-specific CD8+ T cells temporally correlated with the protection elicited by SIVΔnef live attenuated vaccine. PLoS One 2017; 12:e0173929. [PMID: 28333940 PMCID: PMC5363825 DOI: 10.1371/journal.pone.0173929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
The live attenuated vaccine (LAV) SIVmac239Δnef (SIVΔnef) confers the best protection among all the vaccine modalities tested in rhesus macaque model of HIV-1 infection. This vaccine has a unique feature of time-dependent protection: macaques are not protected at 3–5 weeks post vaccination (WPV), whereas immune protection emerges between 15 and 20 WPV. Although the exact mechanisms of the time-dependent protection remain incompletely understood, studies suggested that both cellular and humoral immunities contribute to this time-dependent protection. To further elucidate the mechanisms of protection induced by SIVΔnef, we longitudinally compared the global gene expression profiles of SIV Gag-CM9+ CD8+ (Gag-specific CD8+) T cells from peripheral blood of Mamu-A*01+ rhesus macaques at 3 and 20 WPV using rhesus microarray. We found that gene expression profiles of Gag-specific CD8+ T cells at 20 WPV are qualitatively different from those at 3 WPV. At 20 WPV, the most significant transcriptional changes of Gag-specific CD8+ T cells were genes involved in TCR signaling, differentiation and maturation toward central memory cells, with increased expression of CCR7, TCRα, TCRβ, CD28 and decreased expression of CTLA-4, IFN-γ, RANTES, granzyme A and B. Our study suggests that a higher quality of SIV-specific CD8+ T cells elicited by SIVΔnef over time contributes to the maturation of time-dependent protection.
Collapse
|
10
|
Li S, Folkvord JM, Rakasz EG, Abdelaal HM, Wagstaff RK, Kovacs KJ, Kim HO, Sawahata R, MaWhinney S, Masopust D, Connick E, Skinner PJ. Simian Immunodeficiency Virus-Producing Cells in Follicles Are Partially Suppressed by CD8+ Cells In Vivo. J Virol 2016; 90:11168-11180. [PMID: 27707919 PMCID: PMC5126374 DOI: 10.1128/jvi.01332-16] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/27/2016] [Indexed: 01/24/2023] Open
Abstract
Human immunodeficiency virus (HIV)- and simian immunodeficiency virus (SIV)-specific CD8+ T cells are typically largely excluded from lymphoid B cell follicles, where HIV- and SIV-producing cells are most highly concentrated, indicating that B cell follicles are somewhat of an immunoprivileged site. To gain insights into virus-specific follicular CD8+ T cells, we determined the location and phenotype of follicular SIV-specific CD8+ T cells in situ, the local relationship of these cells to Foxp3+ cells, and the effects of CD8 depletion on levels of follicular SIV-producing cells in chronically SIV-infected rhesus macaques. We found that follicular SIV-specific CD8+ T cells were able to migrate throughout follicular areas, including germinal centers. Many expressed PD-1, indicating that they may have been exhausted. A small subset was in direct contact with and likely inhibited by Foxp3+ cells, and a few were themselves Foxp3+ In addition, subsets of follicular SIV-specific CD8+ T cells expressed low to medium levels of perforin, and subsets were activated and proliferating. Importantly, after CD8 depletion, the number of SIV-producing cells increased in B cell follicles and extrafollicular areas, suggesting that follicular and extrafollicular CD8+ T cells have a suppressive effect on SIV replication. Taken together, these results suggest that during chronic SIV infection, despite high levels of exhaustion and likely inhibition by Foxp3+ cells, a subset of follicular SIV-specific CD8+ T cells are functional and suppress viral replication in vivo These findings support HIV cure strategies that augment functional follicular virus-specific CD8+ T cells to enhance viral control. IMPORTANCE HIV- and SIV-specific CD8+ T cells are typically largely excluded from lymphoid B cell follicles, where virus-producing cells are most highly concentrated, suggesting that B cell follicles are somewhat of an immunoprivileged site where virus-specific CD8+ T cells are not able to clear all follicular HIV- and SIV-producing cells. To gain insights into follicular CD8+ T cell function, we characterized follicular virus-specific CD8+ T cells in situ by using an SIV-infected rhesus macaque model of HIV. We found that subsets of follicular SIV-specific CD8+ T cells are able to migrate throughout the follicle, are likely inhibited by Foxp3+ cells, and are likely exhausted but that, nonetheless, subsets are likely functional, as they express markers consistent with effector function and show signs of suppressing viral replication in vivo These findings support HIV cure strategies that increase the frequency of functional follicular virus-specific CD8+ T cells.
Collapse
Affiliation(s)
- Shengbin Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| | - Joy M Folkvord
- Division of Infectious Diseases, University of Arizona, Tucson, Arizona, USA
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hadia M Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
- Department of Microbiology and Immunology, Zagazig University, Zagazig, Egypt
| | - Reece K Wagstaff
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| | - Katalin J Kovacs
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| | - Hyeon O Kim
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| | - Ryoko Sawahata
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| | - Samantha MaWhinney
- Department of Biostatistics and Informatics, University of Colorado Denver, Aurora, Colorado, USA
| | - David Masopust
- Department of Microbiology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, Arizona, USA
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
11
|
Affiliation(s)
- Akiko Iwasaki
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520;
| |
Collapse
|
12
|
Tjernlund A, Burgener A, Lindvall JM, Peng T, Zhu J, Öhrmalm L, Picker LJ, Broliden K, McElrath MJ, Corey L. In Situ Staining and Laser Capture Microdissection of Lymph Node Residing SIV Gag-Specific CD8+ T cells--A Tool to Interrogate a Functional Immune Response Ex Vivo. PLoS One 2016; 11:e0149907. [PMID: 26986062 PMCID: PMC4795610 DOI: 10.1371/journal.pone.0149907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/05/2016] [Indexed: 11/18/2022] Open
Abstract
While a plethora of data describes the essential role of systemic CD8+ T cells in the control of SIV replication little is known about the local in situ CD8+ T cell immune responses against SIV at the intact tissue level, due to technical limitations. In situ staining, using GagCM9 Qdot 655 multimers, were here combined with laser capture microdissection to detect and collect SIV Gag CM9 specific CD8+ T cells in lymph node tissue from SIV infected rhesus macaques. CD8+ T cells from SIV infected and uninfected rhesus macaques were also collected and compared to the SIV GagCM9 specific CD8+ T cells. Illumina bead array and transcriptional analyses were used to assess the transcriptional profiles and the three different CD8+ T cell populations displayed unique transcriptional patterns. This pilot study demonstrates that rapid and specific immunostaining combined with laser capture microdissection in concert with transcriptional profiling may be used to elucidate phenotypic differences between CD8+ T cells in SIV infection. Such technologies may be useful to determine differences in functional activities of HIV/SIV specific T cells.
Collapse
Affiliation(s)
- Annelie Tjernlund
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:01, 17176 Stockholm, Sweden
- * E-mail:
| | - Adam Burgener
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:01, 17176 Stockholm, Sweden
- National Laboratory for HIV Immunology, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, 730 William Ave. Winnipeg, MB, Canada
| | - Jessica M. Lindvall
- Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Stockholm, Sweden
| | - Tao Peng
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Jia Zhu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America
| | - Lars Öhrmalm
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:01, 17176 Stockholm, Sweden
| | - Louis J. Picker
- Department of Pathology, Vaccine and Gene Therapy Institute, and the Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States of America
| | - Kristina Broliden
- Department of Medicine Solna, Unit of Infectious Diseases, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:01, 17176 Stockholm, Sweden
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
13
|
Iijima N, Iwasaki A. Tissue instruction for migration and retention of TRM cells. Trends Immunol 2015; 36:556-64. [PMID: 26282885 PMCID: PMC4567393 DOI: 10.1016/j.it.2015.07.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/09/2015] [Accepted: 07/13/2015] [Indexed: 01/21/2023]
Abstract
During infection, a subset of effector T cells seeds the lymphoid and non-lymphoid tissues and gives rise to tissue-resident memory T cells (TRM). Recent findings have provided insight into the molecular and cellular mechanisms underlying tissue instruction of TRM cell homing, as well as the programs involved in their retention and maintenance. We review these findings here, highlighting both common features and distinctions between CD4 TRM and CD8 TRM cells. In this context we examine the role of memory lymphocyte clusters (MLCs), and propose that the MLCs serve as an immediate response center consisting of TRM cells on standby, capable of detecting incoming pathogens and mounting robust local immune responses to contain and limit the spread of infectious agents.
Collapse
Affiliation(s)
- Norifumi Iijima
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Akiko Iwasaki
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
14
|
Diem K, Magaret A, Klock A, Jin L, Zhu J, Corey L. Image analysis for accurately counting CD4+ and CD8+ T cells in human tissue. J Virol Methods 2015; 222:117-21. [PMID: 26073660 DOI: 10.1016/j.jviromet.2015.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 11/17/2022]
Abstract
In situ detection of specific cells offers a unique perspective on the spatial interactions between host immune cells and specific viral pathogens or cancers. Most immunohistochemistry techniques require manual cell counting on biopsied and fixed tissue sections. The availability of sophisticated software packages for analyzing fluorescently labeled tissue has made it possible to quickly and accurately quantitate the number of positive cells on such slides. Manual cell counting was compared to automatic cell counting using the program CellProfiler. The two techniques were used to count CD4+ and CD8+ T cells in human genital skin biopsies from herpesvirus type 2 (HSV-2) infected subjects. Manual counting and CellProfiler demonstrated high correlation both in cell counting as well as detection of immune cell "clustering" in tissue, an important visceral component of localized inflammation and characteristic of most chronic infections. Overall, CellProfiler is an effective and accurate method in addition to or replacement of manual cell counting of fluorescently labeled biopsies.
Collapse
Affiliation(s)
- Kurt Diem
- Department of Laboratory Medicine, RR-512 Health Sciences Building, University of Washington, Box 356420, 1959 NE Pacific Street, Seattle, WA 98195, USA.
| | - Amalia Magaret
- Department of Laboratory Medicine, RR-512 Health Sciences Building, University of Washington, Box 356420, 1959 NE Pacific Street, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, 1100 Fairview Avenue N, PO Box 19024, Seattle, WA 98109, USA
| | - Alexis Klock
- Department of Laboratory Medicine, RR-512 Health Sciences Building, University of Washington, Box 356420, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Lei Jin
- Department of Laboratory Medicine, RR-512 Health Sciences Building, University of Washington, Box 356420, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Jia Zhu
- Department of Laboratory Medicine, RR-512 Health Sciences Building, University of Washington, Box 356420, 1959 NE Pacific Street, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, 1100 Fairview Avenue N, PO Box 19024, Seattle, WA 98109, USA
| | - Lawrence Corey
- Department of Laboratory Medicine, RR-512 Health Sciences Building, University of Washington, Box 356420, 1959 NE Pacific Street, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, 1100 Fairview Avenue N, PO Box 19024, Seattle, WA 98109, USA
| |
Collapse
|
15
|
Deruaz M, Luster AD. Chemokine-mediated immune responses in the female genital tract mucosa. Immunol Cell Biol 2015; 93:347-54. [PMID: 25776842 DOI: 10.1038/icb.2015.20] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 01/28/2015] [Accepted: 01/28/2015] [Indexed: 12/25/2022]
Abstract
The genital tract mucosa is the site where sexually transmitted infections gain entry to the host. The immune response at this site is thus critical to provide innate protection against pathogens that are seen for the very first time as well as provide long-term pathogen-specific immunity, which would be required for an effective vaccine against sexually transmitted infection. A finely regulated immune response is therefore required to provide an effective barrier against pathogens without compromising the capacity of the genital tract to allow for successful conception and fetal development. We review recent developments in our understanding of the immune response in the female genital tract to infectious pathogens, using herpes simplex virus-2, human immunodeficiency virus-1 and Chlamydia trachomatis as examples, with a particular focus on the role of chemokines in orchestrating immune cell migration necessary to achieve effective innate and adaptive immune responses in the female genital tract.
Collapse
Affiliation(s)
- Maud Deruaz
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Billingsley JM, Rajakumar PA, Connole MA, Salisch NC, Adnan S, Kuzmichev YV, Hong HS, Reeves RK, Kang HJ, Li W, Li Q, Haase AT, Johnson RP. Characterization of CD8+ T cell differentiation following SIVΔnef vaccination by transcription factor expression profiling. PLoS Pathog 2015; 11:e1004740. [PMID: 25768938 PMCID: PMC4358830 DOI: 10.1371/journal.ppat.1004740] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/10/2015] [Indexed: 01/03/2023] Open
Abstract
The onset of protective immunity against pathogenic SIV challenge in SIVΔnef-vaccinated macaques is delayed for 15-20 weeks, a process that is related to qualitative changes in CD8+ T cell responses induced by SIVΔnef. As a novel approach to characterize cell differentiation following vaccination, we used multi-target qPCR to measure transcription factor expression in naïve and memory subsets of CD8++ T cells, and in SIV-specific CD8+ T cells obtained from SIVΔnef-vaccinated or wild type SIVmac239-infected macaques. Unsupervised clustering of expression profiles organized naïve and memory CD8+ T cells into groups concordant with cell surface phenotype. Transcription factor expression patterns in SIV-specific CD8+ T cells in SIVΔnef-vaccinated animals were distinct from those observed in purified CD8+ T cell subsets obtained from naïve animals, and were intermediate to expression profiles of purified central memory and effector memory T cells. Expression of transcription factors elicited by SIVΔnef vaccination also varied over time: cells obtained at later time points, temporally associated with greater protection, appeared more central-memory like than cells obtained at earlier time points, which appeared more effector memory-like. Expression of transcription factors associated with effector differentiation, such as ID2 and RUNX3, were decreased over time, while expression of transcription factors associated with quiescence or memory differentiation, such as TCF7, BCOR and EOMES, increased. CD8+ T cells specific for a more conserved epitope expressed higher levels of TBX21 and BATF, and appeared more effector-like than cells specific for an escaped epitope, consistent with continued activation by replicating vaccine virus. These data suggest transcription factor expression profiling is a novel method that can provide additional data complementary to the analysis of memory cell differentiation based on classical phenotypic markers. Additionally, these data support the hypothesis that ongoing stimulation by SIVΔnef promotes a distinct protective balance of CD8+ T cell differentiation and activation states. The live attenuated vaccine SIVΔnef can induce robust CD8+ T cell- mediated protection against infection with pathogenic SIV in macaques. Thus, there is substantial interest in characterizing these immune responses to inform HIV vaccine design. Animals challenged at 15–20 weeks post vaccination exhibit robust protection, whereas animals challenged at 5 weeks post-vaccination manifest little protection. Since the frequency of SIV-specific T cells decreases from week 5 to week 20, it is likely that the quality of the response to challenge changes as virus-specific cells differentiate. We applied a novel approach of transcription factor expression profiling to characterize the differences in SIV-specific cell function and phenotype at more protected and less protected time points. Using unsupervised clustering methods informed by expression profiles assessed in purified CD8+ T cell subsets, we show that SIV-specific cells display expression profiles different than any purified CD8+ T cell subset, and intermediate to sorted effector memory and central memory subsets. SIV-specific cells overall appear more effector memory-like at week 5 post-vaccination, and more central memory-like at week 20 post-vaccination. Distinct profiles of CD8+ T cells specific for different SIV epitopes having different immune escape kinetics suggests maturation is regulated by ongoing low-level replication of vaccine virus.
Collapse
Affiliation(s)
- James M. Billingsley
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Premeela A. Rajakumar
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Michelle A. Connole
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - Nadine C. Salisch
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
- Crucell Holland BV, Leiden, The Netherlands
| | - Sama Adnan
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Yury V. Kuzmichev
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - Henoch S. Hong
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - R. Keith Reeves
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Hyung-joo Kang
- Division of Preventive and Behavioral Medicine, University of Massachusetts Medical Center, Worcester, Massachusetts, United States of America
| | - Wenjun Li
- Division of Preventive and Behavioral Medicine, University of Massachusetts Medical Center, Worcester, Massachusetts, United States of America
| | - Qingsheng Li
- Nebraska Center for Virology and School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Ashley T. Haase
- University of Minnesota, Microbiology Department, Minneapolis, Minnesota, United States of America
| | - R. Paul Johnson
- Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
17
|
Fukazawa Y, Lum R, Okoye AA, Park H, Matsuda K, Bae JY, Hagen SI, Shoemaker R, Deleage C, Lucero C, Morcock D, Swanson T, Legasse AW, Axthelm MK, Hesselgesser J, Geleziunas R, Hirsch VM, Edlefsen PT, Piatak M, Estes JD, Lifson JD, Picker LJ. B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in elite controllers. Nat Med 2015; 21:132-9. [PMID: 25599132 PMCID: PMC4320022 DOI: 10.1038/nm.3781] [Citation(s) in RCA: 405] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/30/2014] [Indexed: 12/17/2022]
Abstract
Chronic-phase HIV and simian immunodeficiency virus (SIV) replication is reduced by as much as 10,000-fold in elite controllers (ECs) compared with typical progressors (TPs), but sufficient viral replication persists in EC tissues to allow viral sequence evolution and induce excess immune activation. Here we show that productive SIV infection in rhesus monkey ECs, but not TPs, is markedly restricted to CD4(+) follicular helper T (TFH) cells, suggesting that these EC monkeys' highly effective SIV-specific CD8(+) T cells can effectively clear productive SIV infection from extrafollicular sites, but their relative exclusion from B cell follicles prevents their elimination of productively infected TFH cells. CD8(+) lymphocyte depletion in EC monkeys resulted in a dramatic re-distribution of productive SIV infection to non-TFH cells, with restriction of productive infection to TFH cells resuming upon CD8(+) T cell recovery. Thus, B cell follicles constitute 'sanctuaries' for persistent SIV replication in the presence of potent anti-viral CD8(+) T cell responses, potentially complicating efforts to cure HIV infection with therapeutic vaccination or T cell immunotherapy.
Collapse
Affiliation(s)
- Yoshinori Fukazawa
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Richard Lum
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Afam A. Okoye
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Haesun Park
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Kenta Matsuda
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jin Young Bae
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Shoko I. Hagen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Carissa Lucero
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - David Morcock
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Tonya Swanson
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Alfred W. Legasse
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | | | | | - Vanessa M. Hirsch
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Paul T. Edlefsen
- Statistical Center for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland 21702, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| |
Collapse
|
18
|
Abdelaal HM, Kim HO, Wagstaff R, Sawahata R, Southern PJ, Skinner PJ. Comparison of Vibratome and Compresstome sectioning of fresh primate lymphoid and genital tissues for in situ MHC-tetramer and immunofluorescence staining. Biol Proced Online 2015; 17:2. [PMID: 25657614 PMCID: PMC4318225 DOI: 10.1186/s12575-014-0012-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/29/2014] [Indexed: 11/10/2022] Open
Abstract
Background For decades, the Vibratome served as a standard laboratory resource for sectioning fresh and fixed tissues. In skilled hands, high quality and consistent fresh unfixed tissue sections can be produced using a Vibratome but the sectioning procedure is extremely time consuming. In this study, we conducted a systematic comparison between the Vibratome and a new approach to section fresh unfixed tissues using a Compresstome. We used a Vibratome and a Compresstome to cut fresh unfixed lymphoid and genital non-human primate tissues then used in situ tetramer staining to label virus-specific CD8 T cells and immunofluorescent counter-staining to label B and T cells. We compared the Vibratome and Compresstome in five different sectioning parameters: speed of cutting, chilling capability, specimen stabilization, size of section, and section/staining quality. Results Overall, the Compresstome and Vibratome both produced high quality sections from unfixed spleen, lymph node, vagina, cervix, and uterus, and subsequent immunofluorescent staining was equivalent. The Compresstome however, offered distinct advantages; producing sections approximately 5 times faster than the Vibratome, cutting tissue sections more easily, and allowing production of larger sections. Conclusions A Compresstome can be used to generate fresh unfixed primate lymph node, spleen, vagina, cervix and uterus sections, and is superior to a Vibratome in cutting these fresh tissues.
Collapse
Affiliation(s)
- Hadia M Abdelaal
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA ; Departments of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519 Egypt
| | - Hyeon O Kim
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| | - Reece Wagstaff
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| | - Ryoko Sawahata
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| | - Peter J Southern
- Departments of Microbiology, MMC 196, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455 USA
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, college of veterinary medicine, University of Minnesota, St. Paul, 1971 Commonwealth Avenue, Minnesota, MN 55108 USA
| |
Collapse
|
19
|
Li Q, Zeng M, Duan L, Voss JE, Smith AJ, Pambuccian S, Shang L, Wietgrefe S, Southern PJ, Reilly CS, Skinner PJ, Zupancic ML, Carlis JV, Piatak M, Waterman D, Reeves RK, Masek-Hammerman K, Derdeyn CA, Alpert MD, Evans DT, Kohler H, Müller S, Robinson J, Lifson JD, Burton DR, Johnson RP, Haase AT. Live simian immunodeficiency virus vaccine correlate of protection: local antibody production and concentration on the path of virus entry. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:3113-25. [PMID: 25135832 PMCID: PMC4157131 DOI: 10.4049/jimmunol.1400820] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We sought design principles for a vaccine to prevent HIV transmission to women by identifying correlates of protection conferred by a highly effective live attenuated SIV vaccine in the rhesus macaque animal model. We show that SIVmac239Δnef vaccination recruits plasma cells and induces ectopic lymphoid follicle formation beneath the mucosal epithelium in the rhesus macaque female reproductive tract. The plasma cells and ectopic follicles produce IgG Abs reactive with viral envelope glycoprotein gp41 trimers, and these Abs are concentrated on the path of virus entry by the neonatal FcR in cervical reserve epithelium and in vaginal epithelium. This local Ab production and delivery system correlated spatially and temporally with the maturation of local protection against high-dose pathogenic SIV vaginal challenge. Thus, designing vaccines to elicit production and concentration of Abs at mucosal frontlines could aid in the development of an effective vaccine to protect women against HIV-1.
Collapse
Affiliation(s)
- Qingsheng Li
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Ming Zeng
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Lijie Duan
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - James E Voss
- Department of Immunology and Microbial Science, International AIDS Vaccine Initiative Neutralizing Antibody Center, and Center for HIV/AIDS Vaccine Immunology and Immunogen Design, The Scripps Research Institute, La Jolla, CA 92037; Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA 02129
| | - Anthony J Smith
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Stefan Pambuccian
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Liang Shang
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Stephen Wietgrefe
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Peter J Southern
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - Cavan S Reilly
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN 55455
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Mary L Zupancic
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455
| | - John V Carlis
- Department of Computer Science and Engineering, College of Science and Engineering, University of Minnesota, Minneapolis, MN 55455
| | - Michael Piatak
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., National Cancer Institute, Frederick, MD 21702
| | | | - R Keith Reeves
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772; Infectious Disease Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02115
| | - Katherine Masek-Hammerman
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772; Infectious Disease Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02115
| | - Cynthia A Derdeyn
- Department of Pathology and Laboratory Medicine and Emory Vaccine Center, Emory University, Yerkes, Atlanta, GA 30329
| | - Michael D Alpert
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - David T Evans
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Heinz Kohler
- Department of Microbiology and Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536
| | | | - James Robinson
- Department of Pediatrics, Center for Infectious Diseases, Tulane University, New Orleans, LA 70112
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., National Cancer Institute, Frederick, MD 21702
| | - Dennis R Burton
- Department of Immunology and Microbial Science, International AIDS Vaccine Initiative Neutralizing Antibody Center, and Center for HIV/AIDS Vaccine Immunology and Immunogen Design, The Scripps Research Institute, La Jolla, CA 92037; Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA 02129
| | - R Paul Johnson
- Ragon Institute of MGH, MIT, and Harvard, Charlestown, MA 02129; New England Primate Research Center, Harvard Medical School, Southborough, MA 01772
| | - Ashley T Haase
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, MN 55455;
| |
Collapse
|
20
|
Tuero I, Robert-Guroff M. Challenges in mucosal HIV vaccine development: lessons from non-human primate models. Viruses 2014; 6:3129-58. [PMID: 25196380 PMCID: PMC4147690 DOI: 10.3390/v6083129] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 12/23/2022] Open
Abstract
An efficacious HIV vaccine is urgently needed to curb the AIDS pandemic. The modest protection elicited in the phase III clinical vaccine trial in Thailand provided hope that this goal might be achieved. However, new approaches are necessary for further advances. As HIV is transmitted primarily across mucosal surfaces, development of immunity at these sites is critical, but few clinical vaccine trials have targeted these sites or assessed vaccine-elicited mucosal immune responses. Pre-clinical studies in non-human primate models have facilitated progress in mucosal vaccine development by evaluating candidate vaccine approaches, developing methodologies for collecting and assessing mucosal samples, and providing clues to immune correlates of protective immunity for further investigation. In this review we have focused on non-human primate studies which have provided important information for future design of vaccine strategies, targeting of mucosal inductive sites, and assessment of mucosal immunity. Knowledge gained in these studies will inform mucosal vaccine design and evaluation in human clinical trials.
Collapse
Affiliation(s)
- Iskra Tuero
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Marjorie Robert-Guroff
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|