Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
2.
Nat Immunol ; 25(8): 1411-1421, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38997431

RÉSUMÉ

A subset of individuals exposed to Mycobacterium tuberculosis (Mtb) that we refer to as 'resisters' (RSTR) show evidence of IFN-γ- T cell responses to Mtb-specific antigens despite serially negative results on clinical testing. Here we found that Mtb-specific T cells in RSTR were clonally expanded, confirming the priming of adaptive immune responses following Mtb exposure. RSTR CD4+ T cells showed enrichment of TH17 and regulatory T cell-like functional programs compared to Mtb-specific T cells from individuals with latent Mtb infection. Using public datasets, we showed that these TH17 cell-like functional programs were associated with lack of progression to active tuberculosis among South African adolescents with latent Mtb infection and with bacterial control in nonhuman primates. Our findings suggested that RSTR may successfully control Mtb following exposure and immune priming and established a set of T cell biomarkers to facilitate further study of this clinical phenotype.


Sujet(s)
Lymphocytes T CD4+ , Mycobacterium tuberculosis , Tuberculose , Mycobacterium tuberculosis/immunologie , Humains , Animaux , Adolescent , Tuberculose/immunologie , Tuberculose/microbiologie , Lymphocytes T CD4+/immunologie , Cellules Th17/immunologie , Femelle , Macaca mulatta , Mâle , Phénotype , Interféron gamma/métabolisme , Interféron gamma/immunologie , Antigènes bactériens/immunologie , Tuberculose latente/immunologie , Tuberculose latente/microbiologie , République d'Afrique du Sud , Jeune adulte , Lymphocytes T régulateurs/immunologie , Adulte
3.
JCI Insight ; 9(9)2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38716731

RÉSUMÉ

T cells are required for protective immunity against Mycobacterium tuberculosis. We recently described a cohort of Ugandan household contacts of tuberculosis cases who appear to "resist" M. tuberculosis infection (resisters; RSTRs) and showed that these individuals harbor IFN-γ-independent T cell responses to M. tuberculosis-specific peptide antigens. However, T cells also recognize nonprotein antigens via antigen-presenting systems that are independent of genetic background, known as donor-unrestricted T cells (DURTs). We used tetramer staining and flow cytometry to characterize the association between DURTs and "resistance" to M. tuberculosis infection. Peripheral blood frequencies of most DURT subsets were comparable between RSTRs and latently infected controls (LTBIs). However, we observed a 1.65-fold increase in frequency of MR1-restricted T (MR1T) cells among RSTRs in comparison with LTBIs. Single-cell RNA sequencing of 18,251 MR1T cells sorted from 8 donors revealed 5,150 clonotypes that expressed a common transcriptional program, the majority of which were private. Sequencing of the T cell receptor α/T cell receptor δ (TCRα/δ) repertoire revealed several DURT clonotypes were expanded among RSTRs, including 2 MR1T clonotypes that recognized mycobacteria-infected cells in a TCR-dependent manner. Overall, our data reveal unexpected donor-specific diversity in the TCR repertoire of human MR1T cells as well as associations between mycobacteria-reactive MR1T clonotypes and resistance to M. tuberculosis infection.


Sujet(s)
Mycobacterium tuberculosis , Humains , Mycobacterium tuberculosis/immunologie , Ouganda , Adulte , Mâle , Antigènes mineurs d'histocompatibilité/immunologie , Antigènes mineurs d'histocompatibilité/génétique , Femelle , Tuberculose/immunologie , Tuberculose/microbiologie , Lymphocytes T/immunologie , Tuberculose latente/immunologie , Tuberculose latente/microbiologie , Clones cellulaires/immunologie , Résistance à la maladie/immunologie , Résistance à la maladie/génétique , Jeune adulte , Antigènes d'histocompatibilité de classe I
4.
J Allergy Clin Immunol ; 152(5): 1247-1260, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37460024

RÉSUMÉ

BACKGROUND: Allergen immunotherapy (AIT) is a well-established disease-modifying therapy for allergic rhinitis, yet the fundamental mechanisms underlying its clinical effect remain inadequately understood. Gauging Response in Allergic Rhinitis to Sublingual and Subcutaneous Immunotherapy was a randomized, double-blind, placebo-controlled trial of individuals allergic to timothy grass who received 2 years of placebo (n = 30), subcutaneous immunotherapy (SCIT) (n = 27), or sublingual immunotherapy (SLIT) (n = 27) and were then followed for 1 additional year. OBJECTIVE: We used yearly biospecimens from the Gauging Response in Allergic Rhinitis to Sublingual and Subcutaneous Immunotherapy study to identify molecular mechanisms of response. METHODS: We used longitudinal transcriptomic profiling of nasal brush and PBMC samples after allergen provocation to uncover airway and systemic expression pathways mediating responsiveness to AIT. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01335139, EudraCT Number: 2010-023536-16. RESULTS: SCIT and SLIT demonstrated similar changes in gene module expression over time. In nasal samples, alterations included downregulation of pathways of mucus hypersecretion, leukocyte migration/activation, and endoplasmic reticulum stress (log2 fold changes -0.133 to -0.640, false discovery rates [FDRs] <0.05). We observed upregulation of modules related to epithelial development, junction formation, and lipid metabolism (log2 fold changes 0.104 to 0.393, FDRs <0.05). In PBMCs, modules related to cellular stress response and type 2 cytokine signaling were reduced by immunotherapy (log2 fold changes -0.611 to -0.828, FDRs <0.05). Expression of these modules was also significantly associated with both Total Nasal Symptom Score and peak nasal inspiratory flow, indicating important links between treatment, module expression, and allergen response. CONCLUSIONS: Our results identify specific molecular responses of the nasal airway impacting barrier function, leukocyte migration activation, and mucus secretion that are affected by both SCIT and SLIT, offering potential targets to guide novel strategies for AIT.


Sujet(s)
Rhinite allergique , Immunothérapie sublinguale , Humains , Transcriptome , Agranulocytes , Pollen , Allergènes , Désensibilisation immunologique/méthodes , Immunothérapie sublinguale/méthodes , Phleum , Injections sous-cutanées , Rhinite allergique/thérapie , Rhinite allergique/traitement médicamenteux
5.
EBioMedicine ; 93: 104678, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37379655

RÉSUMÉ

BACKGROUND: The prevalence of tuberculosis among men who work in the gold mines of South Africa is among the highest in the world, but a fraction of miners demonstrate consistently negative results upon tuberculin skin test (TST) and IFN-γ release assay (IGRA). We hypothesized that these "resisters" (RSTRs) may display unconventional immune signatures of exposure to M. tuberculosis (M.tb). METHODS: In a cohort of RSTRs and matched controls with latent TB infection (LTBI), we profiled the functional breadth of M.tb antigen-specific T cell and antibody responses using multi-parameter flow cytometry and systems serology, respectively. FINDINGS: RSTRs and LTBI controls both exhibited IFN-γ independent T-cell and IgG antibody responses to M.tb-specific antigens ESAT-6 and CFP-10. Antigen-specific antibody Fc galactosylation and sialylation were higher among RSTRs. In a combined T-cell and antibody analysis, M.tb lysate-stimulated TNF secretion by T cells correlated positively with levels of purified protein derivative-specific IgG. A multivariate model of the combined data was able to differentiate RSTR and LTBI subjects. INTERPRETATION: IFN-γ independent immune signatures of exposure to M.tb, which are not detected by approved clinical diagnostics, are readily detectable in an occupational cohort uniquely characterized by intense and long-term infection pressure. Further, TNF may mediate a coordinated response between M.tb-specific T-cells and B-cells. FUNDING: This work was supported by the US National Institutes of Health (R01-AI124348 to Boom, Stein, and Hawn; R01-AI125189 and R01-AI146072 to Seshadri; and 75N93019C00071 to Fortune, Alter, Seshadri, and Boom), the Doris Duke Charitable Foundation (Davies), the Bill & Melinda Gates Foundation (OPP1151836 and OPP1109001 to Hawn; and OPP1151840 to Alter), Mass Life Science Foundation (Fortune), and Good Ventures Fund (Fortune).


Sujet(s)
Mycobacterium tuberculosis , Tuberculose , Mâle , Humains , République d'Afrique du Sud/épidémiologie , Tuberculose/diagnostic , Tuberculose/épidémiologie , Antigènes bactériens , Interféron gamma , Test tuberculinique
6.
Front Immunol ; 13: 834757, 2022.
Article de Anglais | MEDLINE | ID: mdl-35432299

RÉSUMÉ

Mycobacterium bovis bacille Calmette-Guérin (BCG) has been used for 100 years and prevents disseminated tuberculosis and death in young children. However, it shows only partial efficacy against pulmonary tuberculosis (TB) in adults, so new vaccines are urgently needed. The protective efficacy of BCG depends on T cells, which are typically activated by pathogen-derived protein antigens that bind to highly polymorphic major histocompatibility complex (MHC) molecules. Some T cells recognize non-protein antigens via antigen presenting systems that are independent of genetic background, leading to their designation as donor-unrestricted T (DURT) cells. Whether live whole cell vaccines, like BCG, can induce durable expansions of DURT cells in humans is not known. We used combinatorial tetramer staining, multi-parameter flow cytometry, and immunosequencing to comprehensively characterize the effect of BCG on activation and expansion of DURT cell subsets. We examined peripheral blood mononuclear cells (PBMC) derived from a Phase I study of South African adults in which samples were archived at baseline, 3 weeks, and 52 weeks post-BCG revaccination. We did not observe a change in the frequency of total mucosal-associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells, germline encoded mycolyl-reactive (GEM) T cells, or γδ T cells at 52 weeks post-BCG. However, immunosequencing revealed a set of TCR-δ clonotypes that were expanded at 52 weeks post-BCG revaccination. These expanded clones expressed the Vδ2 gene segment and could be further defined on the basis of biochemical similarity into several 'meta-clonotypes' that likely recognize similar epitopes. Our data reveal that BCG vaccination leads to durable expansion of DURT cell clonotypes despite a limited effect on total circulating frequencies in the blood and have implications for defining the immunogenicity of candidate whole cell TB vaccines.


Sujet(s)
Vaccin BCG , Mycobacterium tuberculosis , Adulte , Enfant , Enfant d'âge préscolaire , Humains , Rappel de vaccin , Agranulocytes , Récepteurs aux antigènes des cellules T
7.
J Immunol ; 206(6): 1240-1250, 2021 03 15.
Article de Anglais | MEDLINE | ID: mdl-33536255

RÉSUMÉ

Intradermal vaccination with Mycobacterium bovis bacillus Calmette-Guérin (BCG) protects infants from disseminated tuberculosis, and i.v. BCG protects nonhuman primates (NHP) against pulmonary and extrapulmonary tuberculosis. In humans and NHP, protection is thought to be mediated by T cells, which typically recognize bacterial peptide Ags bound to MHC proteins. However, during vertebrate evolution, T cells acquired the capacity to recognize lipid Ags bound to CD1a, CD1b, and CD1c proteins expressed on APCs. It is unknown whether BCG induces T cell immunity to mycobacterial lipids and whether CD1-restricted T cells are resident in the lung. In this study, we developed and validated Macaca mulatta (Mamu) CD1b and CD1c tetramers to probe ex vivo phenotypes and functions of T cells specific for glucose monomycolate (GMM), an immunodominant mycobacterial lipid Ag. We discovered that CD1b and CD1c present GMM to T cells in both humans and NHP. We show that GMM-specific T cells are expanded in rhesus macaque blood 4 wk after i.v. BCG, which has been shown to protect NHP with near-sterilizing efficacy upon M. tuberculosis challenge. After vaccination, these T cells are detected at high frequency within bronchoalveolar fluid and express CD69 and CD103, markers associated with resident memory T cells. Thus, our data expand the repertoire of T cells known to be induced by whole cell mycobacterial vaccines, such as BCG, and show that lipid Ag-specific T cells are resident in the lungs, where they may contribute to protective immunity.


Sujet(s)
Antigènes bactériens/immunologie , Vaccin BCG/administration et posologie , Glycolipides/immunologie , Lymphocytes T/immunologie , Tuberculose/prévention et contrôle , Adolescent , Animaux , Antigènes bactériens/métabolisme , Antigènes CD1/métabolisme , Lignée cellulaire , Enfant , Études de cohortes , Modèles animaux de maladie humaine , Femelle , Glycoprotéines/métabolisme , Volontaires sains , Humains , Injections veineuses , Poumon/cytologie , Poumon/immunologie , Poumon/microbiologie , Macaca mulatta , Mâle , Mycobacterium bovis/immunologie , Mycobacterium tuberculosis/immunologie , Culture de cellules primaires , Lymphocytes T/métabolisme , Tuberculose/sang , Tuberculose/immunologie , Tuberculose/microbiologie
8.
JCI Insight ; 6(6)2021 03 22.
Article de Anglais | MEDLINE | ID: mdl-33621211

RÉSUMÉ

Comorbid medical illnesses, such as obesity and diabetes, are associated with more severe COVID-19, hospitalization, and death. However, the role of the immune system in mediating these clinical outcomes has not been determined. We used multiparameter flow cytometry and systems serology to comprehensively profile the functions of T cells and antibodies targeting spike, nucleocapsid, and envelope proteins in a convalescent cohort of COVID-19 subjects who were either hospitalized (n = 20) or not hospitalized (n = 40). To avoid confounding, subjects were matched by age, sex, ethnicity, and date of symptom onset. Surprisingly, we found that the magnitude and functional breadth of virus-specific CD4+ T cell and antibody responses were consistently higher among hospitalized subjects, particularly those with medical comorbidities. However, an integrated analysis identified more coordination between polyfunctional CD4+ T cells and antibodies targeting the S1 domain of spike among subjects who were not hospitalized. These data reveal a functionally diverse and coordinated response between T cells and antibodies targeting SARS-CoV-2, which is reduced in the presence of comorbid illnesses that are known risk factors for severe COVID-19.


Sujet(s)
Anticorps antiviraux/physiologie , Lymphocytes T CD4+/physiologie , COVID-19/virologie , Hospitalisation , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus , Virion , Adulte , Sujet âgé , Anticorps neutralisants/métabolisme , Anticorps neutralisants/physiologie , Anticorps antiviraux/métabolisme , Lymphocytes T CD4+/métabolisme , COVID-19/épidémiologie , COVID-19/immunologie , Maladies cardiovasculaires/épidémiologie , Maladies cardiovasculaires/immunologie , Comorbidité , Diabète/épidémiologie , Diabète/immunologie , Femelle , Humains , Immunité humorale , Mâle , Adulte d'âge moyen , Nucléocapside , Indice de gravité de la maladie , Enveloppe virale , Protéines virales , Jeune adulte
9.
medRxiv ; 2020 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-33269369

RÉSUMÉ

Comorbid medical illnesses, such as obesity and diabetes, are associated with more severe COVID-19, hospitalization, and death. However, the role of the immune system in mediating these clinical outcomes has not been determined. We used multi-parameter flow cytometry and systems serology to comprehensively profile the functions of T cells and antibodies targeting spike, nucleocapsid, and envelope proteins in a convalescent cohort of COVID-19 subjects who were either hospitalized (n=20) or not hospitalized (n=40). To avoid confounding, subjects were matched by age, sex, ethnicity, and date of symptom onset. Surprisingly, we found that the magnitude and functional breadth of virus-specific CD4 T cell and antibody responses were consistently higher among hospitalized subjects, particularly those with medical comorbidities. However, an integrated analysis identified more coordination between polyfunctional CD4 T-cells and antibodies targeting the S1 domain of spike among subjects that were not hospitalized. These data reveal a functionally diverse and coordinated response between T cells and antibodies targeting SARS-CoV-2 which is reduced in the presence of comorbid illnesses that are known risk factors for severe COVID-19. Our data suggest that isolated measurements of the magnitudes of spike-specific immune responses are likely insufficient to anticipate vaccine efficacy in high-risk populations.

10.
Nat Med ; 25(7): 1175, 2019 Jul.
Article de Anglais | MEDLINE | ID: mdl-31222179

RÉSUMÉ

In the version of this article originally published, there was an error in the abstract. The word disease should not have been included in the sentence "These individuals were highly exposed to Mtb but tested negative disease by IFN-γ release assay and tuberculin skin test, 'resisting' development of classic LTBI". The sentence should have been "These individuals were highly exposed to Mtb but tested negative by IFN-γ release assay and tuberculin skin test, 'resisting' development of classic LTBI." The error has been corrected in the HTML and PDF versions of this article.

11.
Nat Med ; 25(6): 977-987, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-31110348

RÉSUMÉ

Exposure to Mycobacterium tuberculosis (Mtb) results in heterogeneous clinical outcomes including primary progressive tuberculosis and latent Mtb infection (LTBI). Mtb infection is identified using the tuberculin skin test and interferon-γ (IFN-γ) release assay IGRA, and a positive result may prompt chemoprophylaxis to prevent progression to tuberculosis. In the present study, we report on a cohort of Ugandan individuals who were household contacts of patients with TB. These individuals were highly exposed to Mtb but tested negative disease by IFN-γ release assay and tuberculin skin test, 'resisting' development of classic LTBI. We show that 'resisters' possess IgM, class-switched IgG antibody responses and non-IFN-γ T cell responses to the Mtb-specific proteins ESAT6 and CFP10, immunologic evidence of exposure to Mtb. Compared to subjects with classic LTBI, 'resisters' display enhanced antibody avidity and distinct Mtb-specific IgG Fc profiles. These data reveal a distinctive adaptive immune profile among Mtb-exposed subjects, supporting an expanded definition of the host response to Mtb exposure, with implications for public health and the design of clinical trials.


Sujet(s)
Tuberculose latente/immunologie , Mycobacterium tuberculosis/immunologie , Tuberculose/immunologie , Adulte , Anticorps antibactériens/sang , Antigènes bactériens/immunologie , Marqueurs biologiques/métabolisme , Lymphocytes T CD4+/immunologie , Enfant , Études de cohortes , Femelle , Humains , Interféron gamma/immunologie , Tests de libération d'interféron-gamma , Mâle , Test tuberculinique , Ouganda , Jeune adulte
12.
J Infect Dis ; 220(6): 990-1000, 2019 08 09.
Article de Anglais | MEDLINE | ID: mdl-31058977

RÉSUMÉ

BACKGROUND: Herpes simplex virus 2 (HSV2) causes genital herpes in >400 million persons worldwide. METHODS: We conducted a randomized, double-blinded, placebo-controlled trial of a replication-defective HSV2 vaccine, HSV529. Twenty adults were enrolled in each of 3 serogroups of individuals: those negative for both HSV1 and HSV2 (HSV1-/HSV2-), those positive or negative for HSV1 and positive for HSV2 (HSV1±/HSV2+), and those positive for HSV1 and negative for HSV2 (HSV1+/HSV2-). Sixty participants received vaccine or placebo at 0, 1, and 6 months. The primary end point was the frequency of solicited local and systemic reactions to vaccination. RESULTS: Eighty-nine percent of vaccinees experienced mild-to-moderate solicited injection site reactions, compared with 47% of placebo recipients (95% confidence interval [CI], 12.9%-67.6%; P = .006). Sixty-four percent of vaccinees experienced systemic reactions, compared with 53% of placebo recipients (95% CI, -17.9% to 40.2%; P = .44). Seventy-eight percent of HSV1-/HSV2- vaccine recipients had a ≥4-fold increase in neutralizing antibody titer after 3 doses of vaccine, whereas none of the participants in the other serogroups had such responses. HSV2-specific CD4+ T-cell responses were detected in 36%, 46%, and 27% of HSV1-/HSV2-, HSV1±/HSV2+, and HSV1+/HSV2- participants, respectively, 1 month after the third dose of vaccine, and CD8+ T-cell responses were detected in 14%, 8%, and 18% of participants, respectively. CONCLUSIONS: HSV529 vaccine was safe and elicited neutralizing antibody and modest CD4+ T-cell responses in HSV-seronegative vaccinees. CLINICAL TRIALS REGISTRATION: NCT01915212.


Sujet(s)
Herpès génital/prévention et contrôle , Herpès/prévention et contrôle , Herpèsvirus humain de type 2/immunologie , Vaccination , Vaccins antiviraux/administration et posologie , Vaccins antiviraux/immunologie , Adulte , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Lymphocytes T CD4+ , Lymphocytes T CD8+ , Méthode en double aveugle , Femelle , Herpès génital/immunologie , Herpès/immunologie , Herpèsvirus humain de type 1/immunologie , Humains , Mâle , Tests de neutralisation , Vaccins antiviraux/usage thérapeutique , Jeune adulte
13.
Immunogenetics ; 71(7): 465-478, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-31123763

RÉSUMÉ

Invariant NKT (iNKT) cells in both humans and non-human primates are activated by the glycolipid antigen, α-galactosylceramide (α-GalCer). However, the extent to which the molecular mechanisms of antigen recognition and in vivo phenotypes of iNKT cells are conserved among primate species has not been determined. Using an evolutionary genetic approach, we found a lack of diversifying selection in CD1 genes over 45 million years of evolution, which stands in stark contrast to the history of the MHC system for presenting peptide antigens to T cells. The invariant T cell receptor (TCR)-α chain was strictly conserved across all seven primate clades. Invariant NKT cells from rhesus macaques (Macaca mulatta) bind human CD1D-α-GalCer tetramer and are activated by α-GalCer-loaded human CD1D transfectants. The dominant TCR-ß chain cloned from a rhesus-derived iNKT cell line is nearly identical to that found in the human iNKT TCR, and transduction of the rhesus iNKT TCR into human Jurkat cells show that it is sufficient for binding human CD1D-α-GalCer tetramer. Finally, we used a 20-color flow cytometry panel to probe tissue phenotypes of iNKT cells in a cohort of rhesus macaques. We discovered several tissue-resident iNKT populations that have not been previously described in non-human primates but are known in humans, such as TCR-γδ iNKTs. These data reveal a diversity of iNKT cell phenotypes despite convergent evolution of the genes required for lipid antigen presentation and recognition in humans and non-human primates.


Sujet(s)
Antigènes CD1/génétique , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Primates/génétique , Séquence d'acides aminés , Animaux , Antigènes CD1/métabolisme , Séquence conservée , Évolution moléculaire , Femelle , Humains , Cellules Jurkat , Macaca mulatta/immunologie , Mâle , Phénotype , Primates/immunologie , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/métabolisme
14.
J Immunol Methods ; 458: 44-52, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29684428

RÉSUMÉ

CD1 tetramers loaded with lipid antigens facilitate the identification of rare lipid-antigen specific T cells present in human blood and tissue. Because CD1 proteins are structurally non-polymorphic, these tetramers can be applied to genetically diverse human populations, unlike MHC-I and MHC-II tetramers. However, there are no standardized assays to quantify and characterize lipid antigen-specific T cells present within clinical samples. We incorporated CD1b tetramers loaded with the mycobacterial lipid glucose monomycolate (GMM) into a multi-parameter flow cytometry assay. Using a GMM-specific T-cell line, we demonstrate that the assay is linear, reproducible, repeatable, precise, accurate, and has a limit of detection of approximately 0.007%. Having formally validated this assay, we performed a cross-sectional study of healthy U.S. controls and South African adolescents with and without latent tuberculosis infection (LTBI). We show that GMM-specific T cells are specifically detected in South African subjects with LTBI and not in U.S. healthy controls. This assay can be expanded to include additional tetramers or phenotypic markers to characterize GMM-specific T cells in studies of mycobacterial infection, disease, or vaccination.


Sujet(s)
Antigènes CD1/immunologie , Cytométrie en flux/méthodes , Tuberculose latente/diagnostic , Mycobacterium tuberculosis/isolement et purification , Lymphocytes T/immunologie , Adolescent , Adulte , Antigènes bactériens/immunologie , Antigènes CD1/métabolisme , Techniques de culture cellulaire , Lignée cellulaire , Études transversales , Déterminants antigéniques des lymphocytes T/immunologie , Cytométrie en flux/instrumentation , Glycolipides/composition chimique , Glycolipides/immunologie , Volontaires sains , Humains , Immunogénicité des vaccins , Tuberculose latente/immunologie , Tuberculose latente/microbiologie , Tuberculose latente/prévention et contrôle , Mycobacterium tuberculosis/immunologie , Multimérisation de protéines/immunologie , République d'Afrique du Sud , Lymphocytes T/métabolisme , Vaccins antituberculeux/administration et posologie , Vaccins antituberculeux/immunologie , États-Unis
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE