Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.067
2.
Curr Microbiol ; 81(7): 197, 2024 May 30.
Article En | MEDLINE | ID: mdl-38816607

Identifying and evaluating potential vaccine candidates has become one of the main objectives to combat tuberculosis. Among them, mannosylated Apa antigen from Mycobacterium tuberculosis and the non-mannosylated protein expressed in Escherichia coli, have been studied. Although both proteins can induce a protective response in mice, it has been considered that native protein can be dispensed. In this work, we study the protective response induced by Apa expressed in E. coli and in Streptomyces lividans. The latter, like native is secreted as a double band of 45/47 kDa, however, only its 47 kDa band is mannosylated. Both antigens and BCG were intranasal administrated in mice, and animals were then challenged by aerosol with M. tuberculosis H37Rv. The results showed that both, Apa from S. lividans and E. coli conferred statistically significantly protection to animals compared to controls. The cytokine immune response was studied by an immunoassay after animals' immunization, revealing that Apa from S. lividans induced a statistically significant proliferation of T cell, as well as the expression of IFN-γ, IL-1ß, IL-17 and IL-10. In contrast, non-proliferation was obtained with non-mannosylated protein, but induction of IL-12 and IL-17 was observed. Together, these results demonstrate that both proteins were able to modulate a specific immune response against M. tuberculosis, that could be driven by different mechanisms possibly associated with the presence or not of mannosylation. Furthermore, stimulation of cells from BCG-vaccinated animals with the proteins could be an important tool, to help define the use of a given subunit-vaccine after BCG vaccination.


Administration, Intranasal , Cytokines , Mycobacterium tuberculosis , Streptomyces lividans , Tuberculosis , Animals , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/genetics , Mice , Cytokines/metabolism , Tuberculosis/prevention & control , Tuberculosis/immunology , Streptomyces lividans/genetics , Streptomyces lividans/immunology , Aerosols , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/administration & dosage , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Mice, Inbred BALB C , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/administration & dosage
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 367-372, 2024 Apr.
Article Zh | MEDLINE | ID: mdl-38710520

Toll-like receptor 2 (TLR2) is a pattern recognition receptor expressed on the surface of leukocytes. Various ligands can activate or inhibit TLR2, therefore regulating the inflammation and apoptosis of immune cells. Mycobacterium tuberculosis (MTB) typically parasitizes macrophages. Further, after infecting the body, MTB can interact with TLR2 on the surface of various immune cells, including macrophages, leading to the release of cytokines that can affect the state and proliferation of MTB in the body. Additional research is needed to understand the polymorphism of TLR2 at the molecular level. Current studies indicate that the majority of TLR2 polymorphisms are not associated with susceptibility to MTB infection. This review provides an overview of the researches related to TLR2 and its ligands, the immune regulation activities of TLR2 following MTB infection, and the association of TLR2 polymorphism with susceptibility to MTB.


Mycobacterium tuberculosis , Toll-Like Receptor 2 , Tuberculosis , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/immunology , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Polymorphism, Genetic , Animals , Genetic Predisposition to Disease
4.
PLoS Pathog ; 20(5): e1012148, 2024 May.
Article En | MEDLINE | ID: mdl-38728367

Previously, we found that Mycobacterium tuberculosis (Mtb) infection in type 2 diabetes mellitus (T2DM) mice enhances inflammatory cytokine production which drives pathological immune responses and mortality. In the current study, using a T2DM Mtb infection mice model, we determined the mechanisms that make T2DM mice alveolar macrophages (AMs) more inflammatory upon Mtb infection. Among various cell death pathways, necroptosis is a major pathway involved in inflammatory cytokine production by T2DM mice AMs. Anti-TNFR1 antibody treatment of Mtb-infected AMs from T2DM mice significantly reduced expression of receptor interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) (necroptosis markers) and IL-6 production. Metabolic profile comparison of Mtb-infected AMs from T2DM mice and Mtb-infected AMs of nondiabetic control mice indicated that 2-ketohexanoic acid and deoxyadenosine monophosphate were significantly abundant, and acetylcholine and pyridoxine (Vitamin B6) were significantly less abundant in T2DM mice AMs infected with Mtb. 2-Ketohexanoic acid enhanced expression of TNFR1, RIPK3, MLKL and inflammatory cytokine production in the lungs of Mtb-infected nondiabetic mice. In contrast, pyridoxine inhibited RIPK3, MLKL and enhanced expression of Caspase 3 (apoptosis marker) in the lungs of Mtb-infected T2DM mice. Our findings demonstrate that metabolic changes in Mtb-infected T2DM mice enhance TNFR1-mediated necroptosis of AMs, which leads to excess inflammation and lung pathology.


Diabetes Mellitus, Type 2 , Mycobacterium tuberculosis , Necroptosis , Animals , Mice , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Mice, Inbred C57BL , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/microbiology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Male , Cytokines/metabolism
5.
PLoS Pathog ; 20(5): e1012205, 2024 May.
Article En | MEDLINE | ID: mdl-38701094

Mycobacterium tuberculosis (Mtb) infects lung myeloid cells, but the specific Mtb-permissive cells and host mechanisms supporting Mtb persistence during chronic infection are incompletely characterized. We report that after the development of T cell responses, CD11clo monocyte-derived cells harbor more live Mtb than alveolar macrophages (AM), neutrophils, and CD11chi monocyte-derived cells. Transcriptomic and functional studies revealed that the lysosome pathway is underexpressed in this highly permissive subset, characterized by less lysosome content, acidification, and proteolytic activity than AM, along with less nuclear TFEB, a regulator of lysosome biogenesis. Mtb infection does not drive lysosome deficiency in CD11clo monocyte-derived cells but promotes recruitment of monocytes that develop into permissive lung cells, mediated by the Mtb ESX-1 secretion system. The c-Abl tyrosine kinase inhibitor nilotinib activates TFEB and enhances lysosome functions of macrophages in vitro and in vivo, improving control of Mtb infection. Our results suggest that Mtb exploits lysosome-poor lung cells for persistence and targeting lysosome biogenesis is a potential host-directed therapy for tuberculosis.


Lysosomes , Macrophages, Alveolar , Monocytes , Mycobacterium tuberculosis , Lysosomes/metabolism , Lysosomes/microbiology , Animals , Monocytes/metabolism , Monocytes/microbiology , Mice , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/metabolism , Lung/microbiology , Lung/metabolism , Mice, Inbred C57BL , Chronic Disease , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Humans , Tuberculosis/microbiology , Tuberculosis/immunology , Tuberculosis/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
6.
Zhonghua Jie He He Hu Xi Za Zhi ; 47(5): 485-489, 2024 May 12.
Article Zh | MEDLINE | ID: mdl-38706074

Programmed cell death 1 (PD-1) and its ligands, PD-L1 and PD-L2, expressed on a variety of immune cells, play multiple regulatory roles in the host immune response to Mycobacterium tuberculosis infection. In this study, we reviewed that the regulatory roles of PD-1/PD-L1, PD-L2 signaling in the host adaptive immune response, such as the innate response of macrophages, and the interaction between T cells and macrophages in response to MTB. In addition, during MTB infection, PD-1/PD-L1, PD-L2 signaling is also involved in the host inflammatory response, as well as the potential roles of PD-1/PD-L1, PD-L2 in the diagnosis and treatment of tuberculosis.


B7-H1 Antigen , Macrophages , Mycobacterium tuberculosis , Programmed Cell Death 1 Ligand 2 Protein , Programmed Cell Death 1 Receptor , Signal Transduction , Tuberculosis , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Mycobacterium tuberculosis/immunology , Macrophages/immunology , Macrophages/metabolism , Immunity, Innate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Adaptive Immunity
7.
Front Immunol ; 15: 1380971, 2024.
Article En | MEDLINE | ID: mdl-38799462

Mycobacterium tuberculosis (Mtb) remains a major threat worldwide, although only a fraction of infected individuals develops tuberculosis (TB). TB susceptibility is shaped by multiple genetic factors, and we performed comparative immunological analysis of two mouse strains to uncover relevant mechanisms underlying susceptibility and resistance. C57BL/6 mice are relatively TB-resistant, whereas I/St mice are prone to develop severe TB, partly due to the MHC-II allelic variant that shapes suboptimal CD4+ T cell receptor repertoire. We investigated the repertoires of lung-infiltrating helper T cells and B cells at the progressed stage in both strains. We found that lung CD4+ T cell repertoires of infected C57BL/6 but not I/St mice contained convergent TCR clusters with functionally confirmed Mtb specificity. Transcriptomic analysis revealed a more prominent Th1 signature in C57BL/6, and expression of pro-inflammatory IL-16 in I/St lung-infiltrating helper T cells. The two strains also showed distinct Th2 signatures. Furthermore, the humoral response of I/St mice was delayed, less focused, and dominated by IgG/IgM isotypes, whereas C57BL/6 mice generated more Mtb antigen-focused IgA response. We conclude that the inability of I/St mice to produce a timely and efficient anti-Mtb adaptive immune responses arises from a suboptimal helper T cell landscape that also impacts the humoral response, leading to diffuse inflammation and severe disease.


Adaptive Immunity , Genetic Predisposition to Disease , Mice, Inbred C57BL , Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , Mycobacterium tuberculosis/immunology , Adaptive Immunity/genetics , Tuberculosis/immunology , Tuberculosis/genetics , Lung/immunology , Lung/pathology , B-Lymphocytes/immunology , Disease Models, Animal , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
8.
Front Immunol ; 15: 1387454, 2024.
Article En | MEDLINE | ID: mdl-38799468

Introduction: Mycobacteria are known to exert a range of heterologous effects on the immune system. The mycobacteria-based Freund's Complete Adjuvant is a potent non-specific stimulator of the immune response used in immunization protocols promoting antibody production, and Mycobacterium bovis Bacille Calmette Guérin (BCG) vaccination has been linked with decreased morbidity and mortality beyond the specific protection it provides against tuberculosis (TB) in some populations and age groups. The role of heterologous antibodies in this phenomenon, if any, remains unclear and under-studied. Methods: We set out to evaluate antibody responses to a range of unrelated pathogens following infection with Mycobacterium tuberculosis (M.tb) and vaccination with BCG or a candidate TB vaccine, MTBVAC, in non-human primates. Results: We demonstrate a significant increase in the titer of antibodies against SARS-CoV-2, cytomegalovirus, Epstein-Barr virus, tetanus toxoid, and respiratory syncytial virus antigens following low-dose aerosol infection with M.tb. The magnitude of some of these responses correlated with TB disease severity. However, vaccination with BCG administered by the intradermal, intravenous or aerosol routes, or intradermal delivery of MTBVAC, did not increase antibody responses against unrelated pathogens. Discussion: Our findings suggest that it is unlikely that heterologous antibodies contribute to the non-specific effects of these vaccines. The apparent dysregulation of B cell responses associated with TB disease warrants further investigation, with potential implications for risk of B cell cancers and novel therapeutic strategies.


BCG Vaccine , Mycobacterium tuberculosis , Tuberculosis , Vaccination , Animals , BCG Vaccine/immunology , BCG Vaccine/administration & dosage , Tuberculosis/immunology , Tuberculosis/prevention & control , Mycobacterium tuberculosis/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Antibodies, Viral/immunology , Antibodies, Viral/blood , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/administration & dosage , Female , Macaca mulatta , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/prevention & control , Immunity, Heterologous , Male
9.
Cell Mol Life Sci ; 81(1): 203, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698289

Nitrogen metabolism of M. tuberculosis is critical for its survival in infected host cells. M. tuberculosis has evolved sophisticated strategies to switch between de novo synthesis and uptake of various amino acids from host cells for metabolic demands. Pyridoxal phosphate-dependent histidinol phosphate aminotransferase-HspAT enzyme is critically required for histidine biosynthesis. HspAT is involved in metabolic synthesis of histidine, phenylalanine, tyrosine, tryptophan, and novobiocin. We showed that M. tuberculosis Rv2231c is a conserved enzyme with HspAT activity. Rv2231c is a monomeric globular protein that contains α-helices and ß-sheets. It is a secretory and cell wall-localized protein that regulates critical pathogenic attributes. Rv2231c enhances the survival and virulence of recombinant M. smegmatis in infected RAW264.7 macrophage cells. Rv2231c is recognized by the TLR4 innate immune receptor and modulates the host immune response by suppressing the secretion of the antibacterial pro-inflammatory cytokines TNF, IL-12, and IL-6. It also inhibits the expression of co-stimulatory molecules CD80 and CD86 along with antigen presenting molecule MHC-I on macrophage and suppresses reactive nitrogen species formation, thereby promoting M2 macrophage polarization. Recombinant M. smegmatis expressing Rv2231c inhibited apoptosis in macrophages, promoting efficient bacterial survival and proliferation, thereby increasing virulence. Our results indicate that Rv2231c is a moonlighting protein that regulates multiple functions of M. tuberculosis pathophysiology to increase its virulence. These mechanistic insights can be used to better understand the pathogenesis of M. tuberculosis and to design strategies for tuberculosis mitigation.


Macrophages , Mycobacterium tuberculosis , Transaminases , Mice , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Animals , RAW 264.7 Cells , Virulence , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Transaminases/metabolism , Transaminases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Mycobacterium smegmatis/pathogenicity , Mycobacterium smegmatis/metabolism , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/enzymology , Cytokines/metabolism , Toll-Like Receptor 4/metabolism , Humans , Immunity, Innate , Host-Pathogen Interactions/immunology , Tuberculosis/immunology , Tuberculosis/microbiology
10.
Front Immunol ; 15: 1347045, 2024.
Article En | MEDLINE | ID: mdl-38756781

It is essential to understand the interactions and relationships between Mycobacterium tuberculosis (Mtb) and macrophages during the infection in order to design host-directed, immunomodulation-dependent therapeutics to control Mtb. We had reported previously that ornithine acetyltransferase (MtArgJ), a crucial enzyme of the arginine biosynthesis pathway of Mtb, is allosterically inhibited by pranlukast (PRK), which significantly reduces bacterial growth. The present investigation is centered on the immunomodulation in the host by PRK particularly the activation of the host's immune response to counteract bacterial survival and pathogenicity. Here, we show that PRK decreased the bacterial burden in the lungs by upregulating the population of pro-inflammatory interstitial macrophages (IMs) and reducing the population of Mtb susceptible alveolar macrophages (AMs), dendritic cells (DCs), and monocytes (MO). Additionally, we deduce that PRK causes the host macrophages to change their metabolic pathway from fatty acid metabolism to glycolytic metabolism around the log phage of bacterial multiplication. Further, we report that PRK reduced tissue injury by downregulating the Ly6C-positive population of monocytes. Interestingly, PRK treatment improved tissue repair and inflammation resolution by increasing the populations of arginase 1 (Arg-1) and Ym1+Ym2 (chitinase 3-like 3) positive macrophages. In summary, our study found that PRK is useful not only for reducing the tubercular burden but also for promoting the healing of the diseased tissue.


Chromones , Disease Models, Animal , Mycobacterium tuberculosis , Animals , Mycobacterium tuberculosis/immunology , Mice , Chromones/pharmacology , Chromones/therapeutic use , Antitubercular Agents/therapeutic use , Antitubercular Agents/pharmacology , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/drug therapy , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , Mice, Inbred C57BL , Female , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/drug therapy , Lung/microbiology , Lung/immunology , Lung/pathology
11.
Commun Biol ; 7(1): 584, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755239

B cells are important in tuberculosis (TB) immunity, but their role in the human lung is understudied. Here, we characterize B cells from lung tissue and matched blood of patients with TB and found they are decreased in the blood and increased in the lungs, consistent with recruitment to infected tissue, where they are located in granuloma associated lymphoid tissue. Flow cytometry and transcriptomics identify multiple B cell populations in the lung, including those associated with tissue resident memory, germinal centers, antibody secretion, proinflammatory atypical B cells, and regulatory B cells, some of which are expanded in TB disease. Additionally, TB lungs contain high levels of Mtb-reactive antibodies, specifically IgM, which promotes Mtb phagocytosis. Overall, these data reveal the presence of functionally diverse B cell subsets in the lungs of patients with TB and suggest several potential localized roles that may represent a target for interventions to promote immunity or mitigate immunopathology.


B-Lymphocytes , Humans , B-Lymphocytes/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Phenotype , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/genetics , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Male , Female , Adult
12.
Nat Commun ; 15(1): 4216, 2024 May 17.
Article En | MEDLINE | ID: mdl-38760394

Antimicrobial peptides (AMPs), ancient scavengers of bacteria, are very poorly induced in macrophages infected by Mycobacterium tuberculosis (M. tuberculosis), but the underlying mechanism remains unknown. Here, we report that L-alanine interacts with PRSS1 and unfreezes the inhibitory effect of PRSS1 on the activation of NF-κB pathway to induce the expression of AMPs, but mycobacterial alanine dehydrogenase (Ald) Rv2780 hydrolyzes L-alanine and reduces the level of L-alanine in macrophages, thereby suppressing the expression of AMPs to facilitate survival of mycobacteria. Mechanistically, PRSS1 associates with TAK1 and disruptes the formation of TAK1/TAB1 complex to inhibit TAK1-mediated activation of NF-κB pathway, but interaction of L-alanine with PRSS1, disables PRSS1-mediated impairment on TAK1/TAB1 complex formation, thereby triggering the activation of NF-κB pathway to induce expression of AMPs. Moreover, deletion of antimicrobial peptide gene ß-defensin 4 (Defb4) impairs the virulence by Rv2780 during infection in mice. Both L-alanine and the Rv2780 inhibitor, GWP-042, exhibits excellent inhibitory activity against M. tuberculosis infection in vivo. Our findings identify a previously unrecognized mechanism that M. tuberculosis uses its own alanine dehydrogenase to suppress host immunity, and provide insights relevant to the development of effective immunomodulators that target M. tuberculosis.


Alanine , Antimicrobial Peptides , Macrophages , Mycobacterium tuberculosis , NF-kappa B , Tuberculosis , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/metabolism , Animals , Mice , NF-kappa B/metabolism , Humans , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Alanine/metabolism , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/genetics , Tuberculosis/microbiology , Tuberculosis/immunology , Alanine Dehydrogenase/metabolism , Alanine Dehydrogenase/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Signal Transduction , Mice, Inbred C57BL , RAW 264.7 Cells , Female
13.
Life Sci Alliance ; 7(7)2024 Jul.
Article En | MEDLINE | ID: mdl-38803236

Neutrophils can be beneficial or deleterious during tuberculosis (TB). Based on the expression of MHC-II and programmed death ligand 1 (PD-L1), we distinguished two functionally and transcriptionally distinct neutrophil subsets in the lungs of mice infected with mycobacteria. Inflammatory [MHC-II-, PD-L1lo] neutrophils produced inflammasome-dependent IL-1ß in the lungs in response to virulent mycobacteria and "accelerated" deleterious inflammation, which was highly exacerbated in IFN-γR-/- mice. Regulatory [MHC-II+, PD-L1hi] neutrophils "brake" inflammation by suppressing T-cell proliferation and IFN-γ production. Such beneficial regulation, which depends on PD-L1, is controlled by IFN-γR signaling in neutrophils. The hypervirulent HN878 strain from the Beijing genotype curbed PD-L1 expression by regulatory neutrophils, abolishing the braking function and driving deleterious hyperinflammation in the lungs. These findings add a layer of complexity to the roles played by neutrophils in TB and may explain the reactivation of this disease observed in cancer patients treated with anti-PD-L1.


B7-H1 Antigen , Inflammation , Interleukin-1beta , Lung , Neutrophils , Tuberculosis , Animals , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Neutrophils/immunology , Neutrophils/metabolism , Mice , Interleukin-1beta/metabolism , Inflammation/immunology , Inflammation/metabolism , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/metabolism , Lung/immunology , Lung/microbiology , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/immunology , Disease Models, Animal , Female , Humans
14.
JCI Insight ; 9(9)2024 May 08.
Article En | MEDLINE | ID: mdl-38716731

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.


Mycobacterium tuberculosis , Humans , Mycobacterium tuberculosis/immunology , Uganda , Adult , Male , Minor Histocompatibility Antigens/immunology , Minor Histocompatibility Antigens/genetics , Female , Tuberculosis/immunology , Tuberculosis/microbiology , T-Lymphocytes/immunology , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , Clone Cells/immunology , Disease Resistance/immunology , Disease Resistance/genetics , Young Adult , Histocompatibility Antigens Class I
15.
Sci Rep ; 14(1): 10375, 2024 05 06.
Article En | MEDLINE | ID: mdl-38710737

Tuberculosis (TB) a disease caused by Mycobacterium tuberculosis (Mtb) poses a significant threat to human life, and current BCG vaccinations only provide sporadic protection, therefore there is a need for developing efficient vaccines. Numerous immunoinformatic methods have been utilized previously, here for the first time a deep learning framework based on Deconvolutional Neural Networks (DCNN) and Bidirectional Long Short-Term Memory (DCNN-BiLSTM) was used to predict Mtb Multiepitope vaccine (MtbMEV) subunits against six Mtb H37Rv proteins. The trained model was used to design MEV within a few minutes against TB better than other machine learning models with 99.5% accuracy. The MEV has good antigenicity, and physiochemical properties, and is thermostable, soluble, and hydrophilic. The vaccine's BLAST search ruled out the possibility of autoimmune reactions. The secondary structure analysis revealed 87% coil, 10% beta, and 2% alpha helix, while the tertiary structure was highly upgraded after refinement. Molecular docking with TLR3 and TLR4 receptors showed good binding, indicating high immune reactions. Immune response simulation confirmed the generation of innate and adaptive responses. In-silico cloning revealed the vaccine is highly expressed in E. coli. The results can be further experimentally verified using various analyses to establish a candidate vaccine for future clinical trials.


Mycobacterium tuberculosis , Neural Networks, Computer , Tuberculosis Vaccines , Tuberculosis Vaccines/immunology , Mycobacterium tuberculosis/immunology , Humans , Molecular Docking Simulation , Vaccine Development/methods , Epitopes/immunology , Tuberculosis/prevention & control , Tuberculosis/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/chemistry
16.
Proc Natl Acad Sci U S A ; 121(19): e2318003121, 2024 May 07.
Article En | MEDLINE | ID: mdl-38691588

Peptides presented by HLA-E, a molecule with very limited polymorphism, represent attractive targets for T cell receptor (TCR)-based immunotherapies to circumvent the limitations imposed by the high polymorphism of classical HLA genes in the human population. Here, we describe a TCR-based bispecific molecule that potently and selectively binds HLA-E in complex with a peptide encoded by the inhA gene of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis in humans. We reveal the biophysical and structural bases underpinning the potency and specificity of this molecule and demonstrate its ability to redirect polyclonal T cells to target HLA-E-expressing cells transduced with mycobacterial inhA as well as primary cells infected with virulent Mtb. Additionally, we demonstrate elimination of Mtb-infected cells and reduction of intracellular Mtb growth. Our study suggests an approach to enhance host T cell immunity against Mtb and provides proof of principle for an innovative TCR-based therapeutic strategy overcoming HLA polymorphism and therefore applicable to a broader patient population.


Histocompatibility Antigens Class I , Mycobacterium tuberculosis , Receptors, Antigen, T-Cell , T-Lymphocytes , Mycobacterium tuberculosis/immunology , Humans , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , HLA-E Antigens , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Tuberculosis/immunology
17.
Front Immunol ; 15: 1395018, 2024.
Article En | MEDLINE | ID: mdl-38799434

Background: Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), continues to be a major public health problem worldwide. The human immunodeficiency virus (HIV) is another equally important life-threatening pathogen. HIV infection decreases CD4+ T cell levels markedly increasing Mtb co-infections. An appropriate animal model for HIV/Mtb co-infection that can recapitulate the diversity of the immune response in humans during co-infection would facilitate basic and translational research in HIV/Mtb infections. Herein, we describe a novel humanized mouse model. Methods: The irradiated NSG-SGM3 mice were transplanted with human CD34+ hematopoietic stem cells, and the humanization was monitored by staining various immune cell markers for flow cytometry. They were challenged with HIV and/or Mtb, and the CD4+ T cell depletion and HIV viral load were monitored over time. Before necropsy, the live mice were subjected to pulmonary function test and CT scan, and after sacrifice, the lung and spleen homogenates were used to determine Mtb load (CFU) and cytokine/chemokine levels by multiplex assay, and lung sections were analyzed for histopathology. The mouse sera were subjected to metabolomics analysis. Results: Our humanized NSG-SGM3 mice were able to engraft human CD34+ stem cells, which then differentiated into a full-lineage of human immune cell subsets. After co-infection with HIV and Mtb, these mice showed decrease in CD4+ T cell counts overtime and elevated HIV load in the sera, similar to the infection pattern of humans. Additionally, Mtb caused infections in both lungs and spleen, and induced granulomatous lesions in the lungs. Distinct metabolomic profiles were also observed in the tissues from different mouse groups after co-infections. Conclusion: The humanized NSG-SGM3 mice are able to recapitulate the pathogenic effects of HIV and Mtb infections and co-infection at the pathological, immunological and metabolism levels and are therefore a reproducible small animal model for studying HIV/Mtb co-infection.


Coinfection , Disease Models, Animal , HIV Infections , Mycobacterium tuberculosis , Tuberculosis , Animals , Coinfection/immunology , Coinfection/microbiology , HIV Infections/immunology , HIV Infections/complications , Humans , Mice , Tuberculosis/immunology , Mycobacterium tuberculosis/immunology , CD4-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation , Viral Load , HIV-1/immunology , Lung/immunology , Lung/pathology , Lung/virology , Hematopoietic Stem Cells/immunology , Mice, SCID
18.
Sci Rep ; 14(1): 9287, 2024 04 23.
Article En | MEDLINE | ID: mdl-38653771

The Mycobacterium tuberculosis complex (MTBC) comprises nine human-adapted lineages that differ in their geographical distribution. Local adaptation of specific MTBC genotypes to the respective human host population has been invoked in this context. We aimed to assess if bacterial genetics governs MTBC pathogenesis or if local co-adaptation translates into differential susceptibility of human macrophages to infection by different MTBC genotypes. We generated macrophages from cryopreserved blood mononuclear cells of Tanzanian tuberculosis patients, from which the infecting MTBC strains had previously been phylogenetically characterized. We infected these macrophages ex vivo with a phylogenetically similar MTBC strain ("matched infection") or with strains representative of other MTBC lineages ("mismatched infection"). We found that L1 infections resulted in a significantly lower bacterial burden and that the intra-cellular replication rate of L2 strains was significantly higher compared the other MTBC lineages, irrespective of the MTBC lineage originally infecting the patients. Moreover, L4-infected macrophages released significantly greater amounts of TNF-α, IL-6, IL-10, MIP-1ß, and IL-1ß compared to macrophages infected by all other strains. While our results revealed no measurable effect of local adaptation, they further highlight the strong impact of MTBC phylogenetic diversity on the variable outcome of the host-pathogen interaction in human tuberculosis.


Macrophages , Mycobacterium tuberculosis , Phylogeny , Tuberculosis , Humans , Tanzania , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/immunology , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Adult , Male , Female , Genotype
19.
Biochem Biophys Res Commun ; 711: 149920, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38615574

Tuberculosis (TB), a deadly infectious disease induced by Mycobacterium tuberculosis (Mtb), continues to be a global public health issue that kill millions of patents every year. Despite significant efforts have been paid to identify effective TB treatments, the emergence of drug-resistant strains of the disease and the presence of comorbidities in TB patients urges us to explore the detailed mechanisms involved in TB immunity and develop more effective innovative anti-TB strategies. HIF-1α, a protein involved in regulating cellular immune responses during TB infection, has been highlighted as a promising target for the development of novel strategies for TB treatment due to its critical roles in anti-TB host immunity. This review provides a summary of current research progress on the roles of HIF-1α in TB infection, highlighting its importance in regulating the host immune response upon Mtb infection and summarizing the influences and mechanisms of HIF-1α on anti-TB immunological responses of host cells. This review also discusses the various challenges associated with developing HIF-1α as a target for anti-TB therapies, including ensuring specificity and avoiding off-target effects on normal cell function, determining the regulation and expression of HIF-1α in TB patients, and developing drugs that can inhibit HIF-1α. More deep understanding of the molecular mechanisms involved in HIF-1α signaling, its impact on TB host status, and systematic animal testing and clinical trials may benefit the optimization of HIF-1α as a novel therapeutic target for TB.


Antitubercular Agents , Hypoxia-Inducible Factor 1, alpha Subunit , Mycobacterium tuberculosis , Signal Transduction , Tuberculosis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/immunology , Signal Transduction/drug effects , Tuberculosis/drug therapy , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Animals , Antitubercular Agents/therapeutic use , Antitubercular Agents/pharmacology , Molecular Targeted Therapy/methods
20.
Sci Rep ; 14(1): 9141, 2024 04 21.
Article En | MEDLINE | ID: mdl-38644371

Tuberculosis remains a large health threat, despite the availability of the tuberculosis vaccine, BCG. As BCG efficacy gradually decreases from adolescence, BCG-Prime and antigen-booster may be an efficient strategy to confer vaccine efficacy. Mycobacterial DNA-binding protein 1 (MDP1, namely Rv2986c, hupB or HU) is a major Mycobacterium tuberculosis protein that induces vaccine-efficacy by co-administration with CpG DNA. To produce MDP1 for booster-vaccine use, we have created recombinant MDP1 produced in both Escherichia coli (eMDP1) and Mycolicibacterium smegmatis (mMDP1), an avirulent rapid-growing mycobacteria. We tested their immunogenicity by checking interferon (IFN)-gamma production by stimulated peripheral blood cells derived from BCG-vaccinated individuals. Similar to native M. tuberculosis MDP1, we observed that most lysin resides in the C-terminal half of mMDP1 are highly methylated. In contrast, eMDP1 had less post-translational modifications and IFN-gamma stimulation. mMDP1 stimulated the highest amount of IFN-gamma production among the examined native M. tuberculosis proteins including immunodominant MPT32 and Antigen 85 complex. MDP1-mediated IFN-gamma production was more strongly enhanced when combined with a new type of CpG DNA G9.1 than any other tested CpG DNAs. Taken together, these results suggest that the combination of mMDP1 and G9.1 possess high potential use for human booster vaccine against tuberculosis.


BCG Vaccine , Bacterial Proteins , DNA-Binding Proteins , Interferon-gamma , Mycobacterium tuberculosis , Protein Processing, Post-Translational , Humans , Interferon-gamma/metabolism , Bacterial Proteins/immunology , BCG Vaccine/immunology , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mycobacterium tuberculosis/immunology , Recombinant Proteins/immunology , Oligodeoxyribonucleotides/pharmacology , Tuberculosis/prevention & control , Tuberculosis/immunology , CpG Islands , Mycobacterium smegmatis/immunology , Mycobacterium smegmatis/metabolism , Escherichia coli/metabolism , Escherichia coli/genetics , Female
...