51
|
Abstract
A major problem in the analysis of vaccine candidates is the lack of any agreed upon surrogates of efficacy, which means that for diseases that depend on a strong T cell response (HIV, TB especially) the only option is to perform an efficacy trial, involving thousands of subjects, enormous costs, and years before the results are known [1]. We also know that T cell responses are an important part of most pathogen responses, and so identifying key T cell response metrics in early vaccine trials would be generally useful. Given our ignorance of what the most important variables are, what would we like to measure and how can this be accomplished, especially given the explosion of new technologies that are available? What follows is a consideration of what should be measured, with the caveat that some of these will be more important than others.
Collapse
Affiliation(s)
- Mark M Davis
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, United States; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States; Immunology Program, Stanford University School of Medicine, Stanford, CA, United States; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
52
|
Yao T, Shooshtari P, Haeryfar SMM. Leveraging Public Single-Cell and Bulk Transcriptomic Datasets to Delineate MAIT Cell Roles and Phenotypic Characteristics in Human Malignancies. Front Immunol 2020; 11:1691. [PMID: 32849590 PMCID: PMC7413026 DOI: 10.3389/fimmu.2020.01691] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are unconventional, innate-like T lymphocytes that recognize vitamin B metabolites of microbial origin among other antigens displayed by the monomorphic molecule MHC class I-related protein 1 (MR1). Abundant in human tissues, reactive to local inflammatory cues, and endowed with immunomodulatory and cytolytic functions, MAIT cells are likely to play key roles in human malignancies. They accumulate in various tumor microenvironments (TMEs) where they often lose some of their functional capacities. However, the potential roles of MAIT cells in anticancer immunity or cancer progression and their significance in shaping clinical outcomes remain largely unknown. In this study, we analyzed publicly available bulk and single-cell tumor transcriptomic datasets to investigate the tissue distribution, phenotype, and prognostic significance of MAIT cells across several human cancers. We found that expanded MAIT cell clonotypes were often shared between the blood, tumor tissue and adjacent healthy tissue of patients with colorectal, hepatocellular, and non-small cell lung carcinomas. Gene expression comparisons between tumor-infiltrating and healthy tissue MAIT cells revealed the presence of activation and/or exhaustion programs within the TMEs of primary hepatocellular and colorectal carcinomas. Interestingly, in basal and squamous cell carcinomas of the skin, programmed cell death-1 (PD-1) blockade upregulated the expression of several effector genes in tumor-infiltrating MAIT cells. We derived a signature comprising stable and specific MAIT cell gene markers across several tissue compartments and cancer types. By applying this signature to estimate MAIT cell abundance in pan-cancer gene expression data, we demonstrate that a heavier intratumoral MAIT cell presence is positively correlated with a favorable prognosis in esophageal carcinoma but predicts poor overall survival in colorectal and squamous cell lung carcinomas. Finally, in colorectal carcinoma and four other cancer types, we found a positive correlation between MR1 expression and estimated MAIT cell abundance. Collectively, our findings indicate that MAIT cells serve important but diverse roles in human cancers. Our work provides useful models and resources that employ gene expression data platforms to enable future studies in the realm of MAIT cell biology.
Collapse
Affiliation(s)
- Tony Yao
- Department of Microbiology and Immunology, Western University, London, ON, Canada
| | - Parisa Shooshtari
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada.,Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, ON, Canada.,Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada.,Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, ON, Canada.,Division of General Surgery, Department of Surgery, Western University, London, ON, Canada.,Centre for Human Immunology, Western University, London, ON, Canada
| |
Collapse
|
53
|
Hanson AL, Nel HJ, Bradbury L, Phipps J, Thomas R, Lê Cao KA, Kenna TJ, Brown MA. Altered Repertoire Diversity and Disease-Associated Clonal Expansions Revealed by T Cell Receptor Immunosequencing in Ankylosing Spondylitis Patients. Arthritis Rheumatol 2020; 72:1289-1302. [PMID: 32162785 DOI: 10.1002/art.41252] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/05/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Ankylosing spondylitis (AS) is a common spondyloarthropathy primarily affecting the axial skeleton and strongly associated with HLA-B*27 carriage. Genetic evidence implicates both autoinflammatory processes and autoimmunity against an HLA-B*27-restricted autoantigen in immunopathology. In addition to articular symptoms, up to 70% of AS patients present with concurrent bowel inflammation, suggesting that adverse interactions between a genetically primed host immune system and the gut microbiome contribute to the disease. Accordingly, this study aimed to characterize adaptive immune responses to antigenic stimuli in AS. METHODS The peripheral CD4 and CD8 T cell receptor (TCR) repertoire was profiled in AS patients (n = 47) and HLA-B*27-matched healthy controls (n = 38). Repertoire diversity was estimated using the Normalized Shannon Diversity Entropy (NSDE) index, and univariate and multivariate statistical analyses were performed to characterize AS-associated clonal signatures. Furthermore, T cell proliferation and cytokine production in response to immunogenic antigen exposure were investigated in vitro in peripheral blood mononuclear cells from AS patients (n = 19) and HLA-B*27-matched healthy controls (n = 14). RESULTS Based on the NSDE measure of sample diversity across CD4 and CD8 T cell repertoires, AS patients showed increased TCR diversity compared to healthy controls (for CD4 T cells, P = 7.8 × 10-6 ; for CD8 T cells, P = 9.3 × 10-4 ), which was attributed to a significant reduction in the magnitude of peripheral T cell expansions globally. Upon in vitro stimulation, fewer T cells from AS patients than from healthy controls expressed interferon-γ (for CD8 T cells, P = 0.03) and tumor necrosis factor (for CD4 T cells, P = 0.01; for CD8 T cells, P = 0.002). In addition, the CD8 TCR signature was altered in HLA-B*27+ AS patients compared to healthy controls, with significantly expanded Epstein-Barr virus-specific clonotypes (P = 0.03) and cytomegalovirus-specific clonotypes (P = 0.02). HLA-B*27+ AS patients also showed an increased incidence of "public" CD8 TCRs, representing identical clonotypes emerging in response to common antigen encounters, including homologous clonotypes matching those previously isolated from individuals with bacterial-induced reactive arthritis. CONCLUSION The dynamics of peripheral T cell responses in AS patients are altered, suggesting that differential antigen exposure and disrupted adaptive immunity are underlying features of the disease.
Collapse
Affiliation(s)
- Aimee L Hanson
- University of Queensland, Brisbane, Queensland, Australia
| | - Hendrik J Nel
- University of Queensland, Brisbane, Queensland, Australia
| | - Linda Bradbury
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia
| | - Julie Phipps
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia
| | - Ranjeny Thomas
- University of Queensland, Brisbane, Queensland, Australia
| | | | - Tony J Kenna
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia
| | - Matthew A Brown
- Queensland University of Technology and Translational Research Institute, Brisbane, Queensland, Australia, and Guy's and St Thomas' NHS Foundation Trust and King's College London NIHR Biomedical Research Centre, King's College London, UK
| |
Collapse
|
54
|
Pomaznoy M, Kuan R, Lindvall M, Burel JG, Seumois G, Vijayanand P, Taplitz R, Gilman RH, Saito M, Lewinsohn DM, Sette A, Peters B, Lindestam Arlehamn CS. Quantitative and Qualitative Perturbations of CD8 + MAITs in Healthy Mycobacterium tuberculosis-Infected Individuals. Immunohorizons 2020; 4:292-307. [PMID: 32499216 PMCID: PMC7543048 DOI: 10.4049/immunohorizons.2000031] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
CD8 T cells are considered important contributors to the immune response against Mycobacterium tuberculosis, yet limited information is currently known regarding their specific immune signature and phenotype. In this study, we applied a cell population transcriptomics strategy to define immune signatures of human latent tuberculosis infection (LTBI) in memory CD8 T cells. We found a 41-gene signature that discriminates between memory CD8 T cells from healthy LTBI subjects and uninfected controls. The gene signature was dominated by genes associated with mucosal-associated invariant T cells (MAITs) and reflected the lower frequency of MAITs observed in individuals with LTBI. There was no evidence for a conventional CD8 T cell–specific signature between the two cohorts. We, therefore, investigated MAITs in more detail based on Vα7.2 and CD161 expression and staining with an MHC-related protein 1 (MR1) tetramer. This revealed two distinct populations of CD8+Vα7.2+CD161+ MAITs: MR1 tetramer+ and MR1 tetramer−, which both had distinct gene expression compared with memory CD8 T cells. Transcriptomic analysis of LTBI versus noninfected individuals did not reveal significant differences for MR1 tetramer+ MAITs. However, gene expression of MR1 tetramer− MAITs showed large interindividual diversity and a tuberculosis-specific signature. This was further strengthened by a more diverse TCR-α and -β repertoire of MR1 tetramer− cells as compared with MR1 tetramer+. Thus, circulating memory CD8 T cells in subjects with latent tuberculosis have a reduced number of conventional MR1 tetramer+ MAITs as well as a difference in phenotype in the rare population of MR1 tetramer− MAITs compared with uninfected controls.
Collapse
Affiliation(s)
- Mikhail Pomaznoy
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Rebecca Kuan
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Mikaela Lindvall
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Julie G Burel
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Grégory Seumois
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | | | - Randy Taplitz
- Division of Infectious Diseases, University of California San Diego, La Jolla, CA 92093
| | - Robert H Gilman
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205.,Universidad Peruana Caytano Hereida, Lima 15102, Peru
| | - Mayuko Saito
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205.,Department of Virology, Tohuku University Graduate School of Medicine, Sendai 9808575, Japan
| | - David M Lewinsohn
- Department of Medicine, VA Portland Health Care System, Portland, OR 97239.,Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Health & Science University, Portland, OR 97239; and
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037.,Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037.,Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | | |
Collapse
|
55
|
Qiao G, Wang X, Zhou X, Morse MA, Wu J, Wang S, Song Y, Jiang N, Zhao Y, Zhou L, Zhao J, Di Y, Zhu L, Hobeika A, Ren J, Lyerly HK. Immune correlates of clinical benefit in a phase I study of hyperthermia with adoptive T cell immunotherapy in patients with solid tumors. Int J Hyperthermia 2020; 36:74-82. [PMID: 31795830 DOI: 10.1080/02656736.2019.1647350] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose: To characterize the T cell receptor (TCR) repertoire, serum cytokine levels, peripheral blood T lymphocyte populations, safety, and clinical efficacy of hyperthermia (HT) combined with autologous adoptive cell therapy (ACT) and either salvage chemotherapy (CT) or anti-PD-1 antibody in patients with previously treated advanced solid tumors.Materials and methods: Thirty-three (33) patients with ovarian, pancreatic, gastric, colorectal, cervical, or endometrial cancer were recruited into the following therapeutic groups: HT + ACT (n = 10), HT + ACT + anti-PD-1 inhibitor (pembrolizumab) (n = 11) and HT + ACT + CT (n = 12). Peripheral blood was collected to analyze TCR repertoire, measurements of cytokines levels and lymphocyte sub-populations before and after treatment.Results: The objective response rate (ORR) was 30% (10/33), including three complete responses (CR) (9.1%) and seven partial responses (PR) (21.2%) and a disease control rate (DCR = CR + PR + SD) of 66.7% (22 of 33). The most common adverse reactions, blistering, subcutaneous fat induration, local heat-related pain, vomiting and sinus tachycardia, were observed in association with HT. IL-2, IL-4, TNF-α, and IFN-γ levels in peripheral blood were significantly increased among the clinical responders (p < 0.05) while IL-6 and IL-10 were elevated among those with progressive disease (p < 0.05). Peripheral blood CD8+/CD28+ T cells increased (p = 0.002), while the CD4+/CD25+/CD127+Treg cells decreased after therapy (p = 0.012). TCR diversity was substantially increased among the clinical responders.Conclusions: Combining HT with ACT plus either CT or anti-PD-1 antibody was safe, generated clinical responses in previously treated advanced cancers, and promoted TCR repertoire diversity and favorable changes in serum IL-2, IL-4, TNF-α, and IFN-γ levels in clinical responders.
Collapse
Affiliation(s)
- Guoliang Qiao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinna Zhou
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Michael A Morse
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Jiangping Wu
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shuo Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yuguang Song
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ni Jiang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yanjie Zhao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lei Zhou
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jing Zhao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yan Di
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lihong Zhu
- Department of Gynecological Oncology, Beijing Gynecology Hospital, Capital Medical University, Beijing, China
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Jun Ren
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
56
|
Comprehensive analysis of structural and sequencing data reveals almost unconstrained chain pairing in TCRαβ complex. PLoS Comput Biol 2020; 16:e1007714. [PMID: 32163410 PMCID: PMC7093030 DOI: 10.1371/journal.pcbi.1007714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 03/24/2020] [Accepted: 02/04/2020] [Indexed: 11/19/2022] Open
Abstract
Antigen recognition by T-cells is guided by the T-cell receptor (TCR) heterodimer formed by α and β chains. A huge diversity of TCR sequences should be maintained by the immune system in order to be able to mount an effective response towards foreign pathogens, so, due to cooperative binding of α and β chains to the pathogen, any constraints on chain pairing can have a profound effect on immune repertoire structure, diversity and antigen specificity. By integrating available structural data and paired chain sequencing results we were able to show that there are almost no constraints on pairing in TCRαβ complexes, allowing naive T-cell repertoire to reach the highest possible diversity. Additional analysis reveals that the specific choice of contacting amino acids can still have a profound effect on complex conformation. Moreover, antigen-driven selection can distort the uniform landscape of chain pairing, while small, yet significant, differences in the pairing can be attributed to various specialized T-cell subsets such as MAIT and iNKT T-cells, as well as other TCR sets specific to certain antigens. In the present paper we study chain pairing preferences in the T-cell receptor (TCR) heterodimer complex. The TCR molecule is formed by α and β chains and binding of both of these chains to an antigen presented by the major histocompatibility complex (MHC) molecule is required in order to trigger an immune response against foreign pathogens and neoantigens. We show that chain pairing in the TCR complex is nearly random ensuring a highly diverse set of TCRs required for recognition of a vast set of antigens. Our results also show that chain pairing preferences can nevertheless influence TCR complex geometry and biases in TCR chain pairing can be used to identify antigen-driven selection or selection towards specialized subsets of T-cells such as mucosal-associated and natural killer invariant T-cells.
Collapse
|
57
|
Zhang JQ, Siltanen CA, Liu L, Chang KC, Gartner ZJ, Abate AR. Linked optical and gene expression profiling of single cells at high-throughput. Genome Biol 2020; 21:49. [PMID: 32093753 PMCID: PMC7041248 DOI: 10.1186/s13059-020-01958-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Single-cell RNA sequencing has emerged as a powerful tool for characterizing cells, but not all phenotypes of interest can be observed through changes in gene expression. Linking sequencing with optical analysis has provided insight into the molecular basis of cellular function, but current approaches have limited throughput. Here, we present a high-throughput platform for linked optical and gene expression profiling of single cells. We demonstrate accurate fluorescence and gene expression measurements on thousands of cells in a single experiment. We use the platform to characterize DNA and RNA changes through the cell cycle and correlate antibody fluorescence with gene expression. The platform's ability to isolate rare cell subsets and perform multiple measurements, including fluorescence and sequencing-based analysis, holds potential for scalable multi-modal single-cell analysis.
Collapse
Affiliation(s)
- Jesse Q Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California San Francisco, San Francisco, CA, USA
| | - Christian A Siltanen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Leqian Liu
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kai-Chun Chang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Zev J Gartner
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- California Institute for Quantitative Biosciences, University of California San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Adam R Abate
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California San Francisco, San Francisco, CA, USA.
- California Institute for Quantitative Biosciences, University of California San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
58
|
Suliman S, Murphy M, Musvosvi M, Gela A, Meermeier EW, Geldenhuys H, Hopley C, Toefy A, Bilek N, Veldsman A, Hanekom WA, Johnson JL, Boom WH, Obermoser G, Huang H, Hatherill M, Lewinsohn DM, Nemes E, Scriba TJ. MR1-Independent Activation of Human Mucosal-Associated Invariant T Cells by Mycobacteria. THE JOURNAL OF IMMUNOLOGY 2019; 203:2917-2927. [PMID: 31611259 PMCID: PMC6859375 DOI: 10.4049/jimmunol.1900674] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/18/2019] [Indexed: 12/15/2022]
Abstract
Tuberculosis (TB) is the leading cause of mortality from a single infectious agent, Mycobacterium tuberculosis Relevant immune targets of the partially efficacious TB vaccine bacille Calmette-Guérin (BCG) remain poorly defined. Mucosal-associated invariant T (MAIT) cells are MHC-related protein 1 (MR1)-restricted T cells, which are reactive against M. tuberculosis, and underexplored as potential TB vaccine targets. We sought to determine whether BCG vaccination activated mycobacteria-specific MAIT cell responses in humans. We analyzed whole blood samples from M. tuberculosis-infected South African adults who were revaccinated with BCG after a six-month course of isoniazid preventative therapy. In vitro BCG stimulation potently induced IFN-γ expression by phenotypic (CD8+CD26+CD161+) MAIT cells, which constituted the majority (75%) of BCG-reactive IFN-γ-producing CD8+ T cells. BCG revaccination transiently expanded peripheral blood frequencies of BCG-reactive IFN-γ+ MAIT cells, which returned to baseline frequencies a year following vaccination. In another cohort of healthy adults who received BCG at birth, 53% of mycobacteria-reactive-activated CD8 T cells expressed CDR3α TCRs, previously reported as MAIT TCRs, expressing the canonical TRAV1-2-TRAJ33 MAIT TCRα rearrangement. CD26 and CD161 coexpression correlated with TRAV1-2+CD161+ phenotype more accurately in CD8+ than CD4-CD8- MAIT cells. Interestingly, BCG-induced IFN-γ expression by MAIT cells in vitro was mediated by the innate cytokines IL-12 and IL-18 more than MR1-induced TCR signaling, suggesting TCR-independent activation. Collectively, the data suggest that activation of blood MAIT cells by innate inflammatory cytokines is a major mechanism of responsiveness to vaccination with whole cell vaccines against TB or in vitro stimulation with mycobacteria (Clinical trial registration: NCT01119521).
Collapse
Affiliation(s)
- Sara Suliman
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa; .,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.,Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Melissa Murphy
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Anele Gela
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Erin W Meermeier
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239
| | - Hennie Geldenhuys
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Christiaan Hopley
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Asma Toefy
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Ashley Veldsman
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - John L Johnson
- Tuberculosis Research Unit, Case Western Reserve University School of Medicine, Cleveland, OH 44106.,Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - W Henry Boom
- Tuberculosis Research Unit, Case Western Reserve University School of Medicine, Cleveland, OH 44106.,Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Gerlinde Obermoser
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305; and.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Huang Huang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305; and.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - David M Lewinsohn
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|