Your browser doesn't support javascript.
loading
MR1-Independent Activation of Human Mucosal-Associated Invariant T Cells by Mycobacteria.
Suliman, Sara; Murphy, Melissa; Musvosvi, Munyaradzi; Gela, Anele; Meermeier, Erin W; Geldenhuys, Hennie; Hopley, Christiaan; Toefy, Asma; Bilek, Nicole; Veldsman, Ashley; Hanekom, Willem A; Johnson, John L; Boom, W Henry; Obermoser, Gerlinde; Huang, Huang; Hatherill, Mark; Lewinsohn, David M; Nemes, Elisa; Scriba, Thomas J.
Affiliation
  • Suliman S; South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa; ssuliman1@bwh.harvard.edu.
  • Murphy M; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
  • Musvosvi M; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.
  • Gela A; Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.
  • Meermeier EW; South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.
  • Geldenhuys H; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
  • Hopley C; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.
  • Toefy A; South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.
  • Bilek N; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
  • Veldsman A; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.
  • Hanekom WA; South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.
  • Johnson JL; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
  • Boom WH; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.
  • Obermoser G; Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239.
  • Huang H; South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.
  • Hatherill M; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
  • Lewinsohn DM; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.
  • Nemes E; South African Tuberculosis Vaccine Initiative, University of Cape Town, Cape Town 7925, South Africa.
  • Scriba TJ; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
J Immunol ; 203(11): 2917-2927, 2019 12 01.
Article in En | MEDLINE | ID: mdl-31611259
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).
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Histocompatibility Antigens Class I / Minor Histocompatibility Antigens / Mucosal-Associated Invariant T Cells / Mycobacterium tuberculosis Type of study: Etiology_studies / Incidence_studies / Observational_studies / Risk_factors_studies Limits: Adolescent / Child / Humans Language: En Journal: J Immunol Year: 2019 Type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Histocompatibility Antigens Class I / Minor Histocompatibility Antigens / Mucosal-Associated Invariant T Cells / Mycobacterium tuberculosis Type of study: Etiology_studies / Incidence_studies / Observational_studies / Risk_factors_studies Limits: Adolescent / Child / Humans Language: En Journal: J Immunol Year: 2019 Type: Article