Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.281
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 36: 603-638, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29490165

RESUMO

Globally, about 36.7 million people were living with HIV infection at the end of 2015. The most frequent infection co-occurring with HIV-1 is Mycobacterium tuberculosis-374,000 deaths per annum are attributable to HIV-tuberculosis, 75% of those occurring in Africa. HIV-1 infection increases the risk of tuberculosis by a factor of up to 26 and alters its clinical presentation, complicates diagnosis and treatment, and worsens outcome. Although HIV-1-induced depletion of CD4+ T cells underlies all these effects, more widespread immune deficits also contribute to susceptibility and pathogenesis. These defects present a challenge to understand and ameliorate, but also an opportunity to learn and optimize mechanisms that normally protect people against tuberculosis. The most effective means to prevent and ameliorate tuberculosis in HIV-1-infected people is antiretroviral therapy, but this may be complicated by pathological immune deterioration that in turn requires more effective host-directed anti-inflammatory therapies to be derived.


Assuntos
Coinfecção , Infecções por HIV/imunologia , HIV-1/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Terapia Antirretroviral de Alta Atividade , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Progressão da Doença , Variação Genética , Infecções por HIV/diagnóstico , Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/genética , Humanos , Tuberculose/diagnóstico , Tuberculose/microbiologia , Tuberculose/terapia , Replicação Viral
2.
Cell ; 185(25): 4682-4702, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36493751

RESUMO

Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Comunicação Celular , Tuberculose/imunologia
3.
Nat Immunol ; 25(8): 1411-1421, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38997431

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos , Mycobacterium tuberculosis , Tuberculose , Mycobacterium tuberculosis/imunologia , Humanos , Animais , Adolescente , Tuberculose/imunologia , Tuberculose/microbiologia , Linfócitos T CD4-Positivos/imunologia , Células Th17/imunologia , Feminino , Macaca mulatta , Masculino , Fenótipo , Interferon gama/metabolismo , Interferon gama/imunologia , Antígenos de Bactérias/imunologia , Tuberculose Latente/imunologia , Tuberculose Latente/microbiologia , África do Sul , Adulto Jovem , Linfócitos T Reguladores/imunologia , Adulto
4.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33125891

RESUMO

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Assuntos
Células-Tronco Hematopoéticas/microbiologia , Imunidade , Mycobacterium tuberculosis/fisiologia , Mielopoese , Animais , Células da Medula Óssea/metabolismo , Proliferação de Células , Suscetibilidade a Doenças , Homeostase , Interferon Tipo I/metabolismo , Ferro/metabolismo , Cinética , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Necrose , Transdução de Sinais , Transcrição Gênica , Tuberculose/imunologia , Tuberculose/microbiologia , Tuberculose/patologia
5.
Cell ; 181(5): 1016-1035.e19, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32413319

RESUMO

There is pressing urgency to understand the pathogenesis of the severe acute respiratory syndrome coronavirus clade 2 (SARS-CoV-2), which causes the disease COVID-19. SARS-CoV-2 spike (S) protein binds angiotensin-converting enzyme 2 (ACE2), and in concert with host proteases, principally transmembrane serine protease 2 (TMPRSS2), promotes cellular entry. The cell subsets targeted by SARS-CoV-2 in host tissues and the factors that regulate ACE2 expression remain unknown. Here, we leverage human, non-human primate, and mouse single-cell RNA-sequencing (scRNA-seq) datasets across health and disease to uncover putative targets of SARS-CoV-2 among tissue-resident cell subsets. We identify ACE2 and TMPRSS2 co-expressing cells within lung type II pneumocytes, ileal absorptive enterocytes, and nasal goblet secretory cells. Strikingly, we discovered that ACE2 is a human interferon-stimulated gene (ISG) in vitro using airway epithelial cells and extend our findings to in vivo viral infections. Our data suggest that SARS-CoV-2 could exploit species-specific interferon-driven upregulation of ACE2, a tissue-protective mediator during lung injury, to enhance infection.


Assuntos
Células Epiteliais Alveolares/metabolismo , Enterócitos/metabolismo , Células Caliciformes/metabolismo , Interferon Tipo I/metabolismo , Mucosa Nasal/citologia , Peptidil Dipeptidase A/genética , Adolescente , Células Epiteliais Alveolares/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Betacoronavirus/fisiologia , COVID-19 , Linhagem Celular , Células Cultivadas , Criança , Infecções por Coronavirus/virologia , Enterócitos/imunologia , Células Caliciformes/imunologia , Infecções por HIV/imunologia , Humanos , Influenza Humana/imunologia , Interferon Tipo I/imunologia , Pulmão/citologia , Pulmão/patologia , Macaca mulatta , Camundongos , Mycobacterium tuberculosis , Mucosa Nasal/imunologia , Pandemias , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/virologia , Receptores Virais/genética , SARS-CoV-2 , Serina Endopeptidases/metabolismo , Análise de Célula Única , Tuberculose/imunologia , Regulação para Cima
6.
Nat Immunol ; 23(2): 318-329, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35058616

RESUMO

Tuberculosis (TB) in humans is characterized by formation of immune-rich granulomas in infected tissues, the architecture and composition of which are thought to affect disease outcome. However, our understanding of the spatial relationships that control human granulomas is limited. Here, we used multiplexed ion beam imaging by time of flight (MIBI-TOF) to image 37 proteins in tissues from patients with active TB. We constructed a comprehensive atlas that maps 19 cell subsets across 8 spatial microenvironments. This atlas shows an IFN-γ-depleted microenvironment enriched for TGF-ß, regulatory T cells and IDO1+ PD-L1+ myeloid cells. In a further transcriptomic meta-analysis of peripheral blood from patients with TB, immunoregulatory trends mirror those identified by granuloma imaging. Notably, PD-L1 expression is associated with progression to active TB and treatment response. These data indicate that in TB granulomas, there are local spatially coordinated immunoregulatory programs with systemic manifestations that define active TB.


Assuntos
Granuloma/imunologia , Tuberculose/imunologia , Antígeno B7-H1/imunologia , Células Cultivadas , Citocinas/imunologia , Perfilação da Expressão Gênica/métodos , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Pulmão/imunologia , Mycobacterium tuberculosis/imunologia , Células Mieloides/imunologia
7.
Cell ; 178(6): 1344-1361.e11, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31474371

RESUMO

Necrosis of infected macrophages constitutes a critical pathogenetic event in tuberculosis by releasing mycobacteria into the growth-permissive extracellular environment. In zebrafish infected with Mycobacterium marinum or Mycobacterium tuberculosis, excess tumor necrosis factor triggers programmed necrosis of infected macrophages through the production of mitochondrial reactive oxygen species (ROS) and the participation of cyclophilin D, a component of the mitochondrial permeability transition pore. Here, we show that this necrosis pathway is not mitochondrion-intrinsic but results from an inter-organellar circuit initiating and culminating in the mitochondrion. Mitochondrial ROS induce production of lysosomal ceramide that ultimately activates the cytosolic protein BAX. BAX promotes calcium flow from the endoplasmic reticulum into the mitochondrion through ryanodine receptors, and the resultant mitochondrial calcium overload triggers cyclophilin-D-mediated necrosis. We identify ryanodine receptors and plasma membrane L-type calcium channels as druggable targets to intercept mitochondrial calcium overload and necrosis of mycobacterium-infected zebrafish and human macrophages.


Assuntos
Macrófagos/microbiologia , Macrófagos/patologia , Mitocôndrias/metabolismo , Infecções por Mycobacterium não Tuberculosas/metabolismo , Tuberculose/imunologia , Tuberculose/patologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose , Cálcio/metabolismo , Retículo Endoplasmático/microbiologia , Humanos , Lisossomos/microbiologia , Potencial da Membrana Mitocondrial , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium marinum , Mycobacterium tuberculosis , Necrose , Espécies Reativas de Oxigênio/metabolismo , Células THP-1 , Peixe-Zebra
8.
Nat Immunol ; 22(12): 1515-1523, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34811542

RESUMO

Development of an effective tuberculosis (TB) vaccine has suffered from an incomplete understanding of the correlates of protection against Mycobacterium tuberculosis (Mtb). Intravenous (i.v.) vaccination with Bacille Calmette-Guérin (BCG) provides nearly complete protection against TB in rhesus macaques, but the antibody response it elicits remains incompletely defined. Here we show that i.v. BCG drives superior antibody responses in the plasma and the lungs of rhesus macaques compared to traditional intradermal BCG administration. While i.v. BCG broadly expands antibody titers and functions, IgM titers in the plasma and lungs of immunized macaques are among the strongest markers of reduced bacterial burden. IgM was also enriched in macaques that received protective vaccination with an attenuated strain of Mtb. Finally, an Mtb-specific IgM monoclonal antibody reduced Mtb survival in vitro. Collectively, these data highlight the potential importance of IgM responses as a marker and mediator of protection against TB.


Assuntos
Anticorpos Antibacterianos/sangue , Vacina BCG/administração & dosagem , Imunogenicidade da Vacina , Imunoglobulina M/sangue , Mycobacterium tuberculosis/imunologia , Tuberculose/prevenção & controle , Vacinação , Administração Intravenosa , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Macaca mulatta , Mycobacterium tuberculosis/patogenicidade , Fatores de Tempo , Tuberculose/imunologia , Tuberculose/microbiologia
9.
Immunity ; 57(10): 2380-2398.e6, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39214090

RESUMO

Immunological priming-in the context of either prior infection or vaccination-elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrated that prior Mtb infection elicited a long-lasting protective response against subsequent Mtb exposure and was CD4+ T cell dependent. By analyzing data from primary infection, reinfection, and reinfection-CD4+ T cell-depleted granulomas, we found that the presence of CD4+ T cells during reinfection resulted in a less inflammatory lung milieu characterized by reprogrammed CD8+ T cells, reduced neutrophilia, and blunted type 1 immune signaling among myeloid cells. These results open avenues for developing vaccines and therapeutics that not only target lymphocytes but also modulate innate immune cells to limit tuberculosis (TB) disease.


Assuntos
Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Granuloma , Imunomodulação , Mycobacterium tuberculosis , Reinfecção , Animais , Linfócitos T CD4-Positivos/imunologia , Mycobacterium tuberculosis/imunologia , Reinfecção/imunologia , Granuloma/imunologia , Granuloma/microbiologia , Linfócitos T CD8-Positivos/imunologia , Tuberculose/imunologia , Tuberculose/microbiologia , Modelos Animais de Doenças , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Humanos , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia
10.
Immunity ; 57(9): 2095-2107.e8, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39153479

RESUMO

Although the Bacille-Calmette-Guérin (BCG) vaccine is used to prevent tuberculosis, it also offers protection against a diverse range of non-mycobacterial infections. However, the underlying protective mechanisms in humans are not yet fully understood. Here, we surveyed at single-cell resolution the gene expression and chromatin landscape of human bone marrow, aspirated before and 90 days after BCG vaccination or placebo. We showed that BCG alters both the gene expression and epigenetic profiles of human hematopoietic stem and progenitor cells (HSPCs). Changes in gene expression occurred primarily within uncommitted stem cells. By contrast, changes in chromatin accessibility were most prevalent within differentiated progenitor cells at sites influenced by Kruppel-like factor (KLF) and early growth response (EGR) transcription factors and were highly correlated (r > 0.8) with the interleukin (IL)-1ß secretion capacity of paired peripheral blood mononuclear cells (PBMCs). Our findings shed light on BCG vaccination's profound and lasting effects on HSPCs and its influence on innate immune responses and trained immunity.


Assuntos
Vacina BCG , Epigênese Genética , Imunidade Inata , Vacinação , Humanos , Vacina BCG/imunologia , Epigênese Genética/imunologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Interleucina-1beta/metabolismo , Medula Óssea/imunologia , Tuberculose/imunologia , Tuberculose/prevenção & controle , Adulto , Leucócitos Mononucleares/imunologia , Cromatina/metabolismo , Feminino , Masculino , Diferenciação Celular/imunologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/imunologia
11.
Cell ; 172(1-2): 176-190.e19, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29328912

RESUMO

The dogma that adaptive immunity is the only arm of the immune response with memory capacity has been recently challenged by several studies demonstrating evidence for memory-like innate immune training. However, the underlying mechanisms and location for generating such innate memory responses in vivo remain unknown. Here, we show that access of Bacillus Calmette-Guérin (BCG) to the bone marrow (BM) changes the transcriptional landscape of hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs), leading to local cell expansion and enhanced myelopoiesis at the expense of lymphopoiesis. Importantly, BCG-educated HSCs generate epigenetically modified macrophages that provide significantly better protection against virulent M. tuberculosis infection than naïve macrophages. By using parabiotic and chimeric mice, as well as adoptive transfer approaches, we demonstrate that training of the monocyte/macrophage lineage via BCG-induced HSC reprogramming is sustainable in vivo. Our results indicate that targeting the HSC compartment provides a novel approach for vaccine development.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Imunidade Inata , Memória Imunológica , Mycobacterium bovis/imunologia , Transcriptoma , Animais , Linhagem Celular , Células Cultivadas , Epigênese Genética , Hematopoese , Camundongos , Camundongos Endogâmicos C57BL , Tuberculose/imunologia
12.
Nat Immunol ; 21(4): 464-476, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32205882

RESUMO

Although mouse infection models have been extensively used to study the host response to Mycobacterium tuberculosis, their validity in revealing determinants of human tuberculosis (TB) resistance and disease progression has been heavily debated. Here, we show that the modular transcriptional signature in the blood of susceptible mice infected with a clinical isolate of M. tuberculosis resembles that of active human TB disease, with dominance of a type I interferon response and neutrophil activation and recruitment, together with a loss in B lymphocyte, natural killer and T cell effector responses. In addition, resistant but not susceptible strains of mice show increased lung B cell, natural killer and T cell effector responses in the lung upon infection. Notably, the blood signature of active disease shared by mice and humans is also evident in latent TB progressors before diagnosis, suggesting that these responses both predict and contribute to the pathogenesis of progressive M. tuberculosis infection.


Assuntos
Transcriptoma/imunologia , Tuberculose/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/microbiologia , Humanos , Interferon Tipo I/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/imunologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Tuberculose/microbiologia
15.
Cell ; 167(2): 306-307, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27716502

RESUMO

In this issue of Cell, Lu et al. provide important insights on the efficacy of human antibodies to Mycobacterium tuberculosis and on how functional heterogeneity of the antibody response may explain a century of contradictory evidence for the role of humoral immunity in defense against tuberculosis.


Assuntos
Anticorpos Antibacterianos/imunologia , Mycobacterium tuberculosis/imunologia , Humanos , Imunidade Humoral , Imunoglobulinas , Tuberculose/imunologia
16.
Cell ; 165(1): 139-152, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27015311

RESUMO

A zebrafish genetic screen for determinants of susceptibility to Mycobacterium marinum identified a hypersusceptible mutant deficient in lysosomal cysteine cathepsins that manifests hallmarks of human lysosomal storage diseases. Under homeostatic conditions, mutant macrophages accumulate undigested lysosomal material, which disrupts endocytic recycling and impairs their migration to, and thus engulfment of, dying cells. This causes a buildup of unengulfed cell debris. During mycobacterial infection, macrophages with lysosomal storage cannot migrate toward infected macrophages undergoing apoptosis in the tuberculous granuloma. The unengulfed apoptotic macrophages undergo secondary necrosis, causing granuloma breakdown and increased mycobacterial growth. Macrophage lysosomal storage similarly impairs migration to newly infecting mycobacteria. This phenotype is recapitulated in human smokers, who are at increased risk for tuberculosis. A majority of their alveolar macrophages exhibit lysosomal accumulations of tobacco smoke particulates and do not migrate to Mycobacterium tuberculosis. The incapacitation of highly microbicidal first-responding macrophages may contribute to smokers' susceptibility to tuberculosis.


Assuntos
Suscetibilidade a Doenças , Lisossomos/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , Infecções por Mycobacterium/imunologia , Infecções por Mycobacterium/patologia , Animais , Granuloma/metabolismo , Macrófagos/citologia , Macrófagos Alveolares/imunologia , Mycobacterium marinum , Alvéolos Pulmonares/imunologia , Fumar , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vesículas Transportadoras/metabolismo , Tuberculose/imunologia , Tuberculose/patologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
17.
Nat Immunol ; 19(11): 1159-1168, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30333612

RESUMO

Blood transcriptomics analysis of tuberculosis has revealed an interferon-inducible gene signature that diminishes in expression after successful treatment; this promises improved diagnostics and treatment monitoring, which are essential for the eradication of tuberculosis. Sensitive radiography revealing lung abnormalities and blood transcriptomics have demonstrated heterogeneity in patients with active tuberculosis and exposed asymptomatic people with latent tuberculosis, suggestive of a continuum of infection and immune states. Here we describe the immune response to infection with Mycobacterium tuberculosis revealed through the use of transcriptomics, as well as differences among clinical phenotypes of infection that might provide information on temporal changes in host immunity associated with evolving infection. We also review the diverse blood transcriptional signatures, composed of small sets of genes, that have been proposed for the diagnosis of tuberculosis and the identification of at-risk asymptomatic people and suggest novel approaches for the development of such biomarkers for clinical use.


Assuntos
Biomarcadores/sangue , Perfilação da Expressão Gênica/métodos , Tuberculose/imunologia , Humanos , Transcriptoma/imunologia , Tuberculose/sangue , Tuberculose/diagnóstico
18.
Nat Immunol ; 19(9): 973-985, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30127434

RESUMO

Human inborn errors of IFN-γ immunity underlie mycobacterial diseases. We describe patients with Mycobacterium bovis (BCG) disease who are homozygous for loss-of-function mutations of SPPL2A. This gene encodes a transmembrane protease that degrades the N-terminal fragment (NTF) of CD74 (HLA invariant chain) in antigen-presenting cells. The CD74 NTF therefore accumulates in the HLA class II+ myeloid and lymphoid cells of SPPL2a-deficient patients. This toxic fragment selectively depletes IL-12- and IL-23-producing CD1c+ conventional dendritic cells (cDC2s) and their circulating progenitors. Moreover, SPPL2a-deficient memory TH1* cells selectively fail to produce IFN-γ when stimulated with mycobacterial antigens in vitro. Finally, Sppl2a-/- mice lack cDC2s, have CD4+ T cells that produce small amounts of IFN-γ after BCG infection, and are highly susceptible to infection with BCG or Mycobacterium tuberculosis. These findings suggest that inherited SPPL2a deficiency in humans underlies mycobacterial disease by decreasing the numbers of cDC2s and impairing IFN-γ production by mycobacterium-specific memory TH1* cells.


Assuntos
Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Células Dendríticas/imunologia , Proteínas de Membrana/metabolismo , Infecções por Mycobacterium/imunologia , Mycobacterium bovis/fisiologia , Mycobacterium tuberculosis/fisiologia , Células Th1/imunologia , Tuberculose/imunologia , Animais , Antígenos de Diferenciação de Linfócitos B/metabolismo , Células Cultivadas , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunidade , Memória Imunológica , Lactente , Interferon gama/metabolismo , Linfadenopatia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Infecções por Mycobacterium/genética , Vacinação
19.
Immunity ; 54(3): 526-541.e7, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33515487

RESUMO

Chronic viral infections increase severity of Mycobacterium tuberculosis (Mtb) coinfection. Here, we examined how chronic viral infections alter the pulmonary microenvironment to foster coinfection and worsen disease severity. We developed a coordinated system of chronic virus and Mtb infection that induced central clinical manifestations of coinfection, including increased Mtb burden, extra-pulmonary dissemination, and heightened mortality. These disease states were not due to chronic virus-induced immunosuppression or exhaustion; rather, increased amounts of the cytokine TNFα initially arrested pulmonary Mtb growth, impeding dendritic cell mediated antigen transportation to the lymph node and subverting immune-surveillance, allowing bacterial sanctuary. The cryptic Mtb replication delayed CD4 T cell priming, redirecting T helper (Th) 1 toward Th17 differentiation and increasing pulmonary neutrophilia, which diminished long-term survival. Temporally restoring CD4 T cell induction overcame these diverse disease sequelae to enhance Mtb control. Thus, Mtb co-opts TNFα from the chronic inflammatory environment to subvert immune-surveillance, avert early immune function, and foster long-term coinfection.


Assuntos
Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Mycobacterium tuberculosis/fisiologia , Neutrófilos/imunologia , Células Th1/imunologia , Células Th17/imunologia , Tuberculose/imunologia , Imunidade Adaptativa , Animais , Doença Crônica , Coinfecção , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagocitose , Índice de Gravidade de Doença , Fatores de Tempo
20.
Cell ; 159(7): 1497-509, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25525872

RESUMO

Tuberculosis, an ancient disease of mankind, remains one of the major infectious causes of human death. We examine newly discovered facets of tuberculosis pathogenesis and explore the evolution of its causative organism Mycobacterium tuberculosis from soil dweller to human pathogen. M. tuberculosis has coevolved with the human host to evade and exploit host macrophages and other immune cells in multiple ways. Though the host can often clear infection, the organism can cause transmissible disease in enough individuals to sustain itself. Tuberculosis is a near-perfect paradigm of a host-pathogen relationship, and that may be the challenge to the development of new therapies for its eradication.


Assuntos
Evasão da Resposta Imune , Mycobacterium tuberculosis/imunologia , Tuberculose/microbiologia , Animais , Granuloma/imunologia , Granuloma/microbiologia , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Tuberculose/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA