Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 7(1): 294, 2024 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-38461214

RESUMO

The continuing emergence of new strains of antibiotic-resistant bacteria has renewed interest in phage therapy; however, there has been limited progress in applying phage therapy to multi-drug resistant Mycobacterium tuberculosis (Mtb) infections. In this study, we show that bacteriophage strains D29 and DS6A can efficiently lyse Mtb H37Rv in 7H10 agar plates. However, only phage DS6A efficiently kills H37Rv in liquid culture and in Mtb-infected human primary macrophages. We further show in subsequent experiments that, after the humanized mice were infected with aerosolized H37Rv, then treated with DS6A intravenously, the DS6A treated mice showed increased body weight and improved pulmonary function relative to control mice. Furthermore, DS6A reduces Mtb load in mouse organs with greater efficacy in the spleen. These results demonstrate the feasibility of developing phage therapy as an effective therapeutic against Mtb infection.


Assuntos
Mycobacterium tuberculosis , Terapia por Fagos , Tuberculose , Animais , Camundongos , Humanos , Tuberculose/terapia , Tuberculose/microbiologia , Macrófagos/microbiologia
2.
PLoS Pathog ; 18(4): e1010454, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35363832

RESUMO

Nontuberculous mycobacteria (NTM) infection is common in patients with structural lung damage. To address how NTM infection is established and causes lung damage, we established an NTM mouse model by intranasal inoculation of clinical isolates of M. intracellulare. During the 39-week course of infection, the bacteria persistently grew in the lung and caused progressive granulomatous and fibrotic lung damage with mortality exceeding 50%. Lung neutrophils were significantly increased at 1 week postinfection, reduced at 2 weeks postinfection and increased again at 39 weeks postinfection. IL-17A was increased in the lungs at 1-2 weeks of infection and reduced at 3 weeks postinfection. Depletion of neutrophils during early (0-2 weeks) and late (32-34 weeks) infection had no effect on mortality or lung damage in chronically infected mice. However, neutralization of IL-17A during early infection significantly reduced bacterial burden, fibrotic lung damage, and mortality in chronically infected mice. Since it is known that IL-17A regulates matrix metalloproteinases (MMPs) and that MMPs contribute to the pathogenesis of pulmonary fibrosis, we determined the levels of MMPs in the lungs of M. intracellulare-infected mice. Interestingly, MMP-3 was significantly reduced by anti-IL-17A neutralizing antibody. Moreover, in vitro data showed that exogenous IL-17A exaggerated the production of MMP-3 by lung epithelial cells upon M. intracellulare infection. Collectively, our findings suggest that early IL-17A production precedes and promotes organized pulmonary M. intracellulare infection in mice, at least in part through MMP-3 production.


Assuntos
Infecção por Mycobacterium avium-intracellulare , Animais , Humanos , Interleucina-17 , Pulmão , Metaloproteinase 3 da Matriz , Camundongos , Infecção por Mycobacterium avium-intracellulare/microbiologia , Infecção por Mycobacterium avium-intracellulare/patologia
3.
Mol Immunol ; 135: 285-293, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33957478

RESUMO

Despite its critical roles in immune responses against tuberculosis infection and immune pathology, the molecular details of interleukin (IL)-1ß production in tuberculosis infection remain elusive. To explore IL-1ß production in tuberculosis infection, we infected mouse bone marrow-derived macrophages (BMDM) with Mycobacterium tuberculosis (Mtb) H37Rv, its early secreted antigenic target protein of 6 kDa (ESAT-6) gene deletion (H37Rv:Δ3875) or complemented strain (H37Rv:Δ3875C) and evaluated IL-1ß production. H37Rv induced significantly increased IL-1ß production by BMDMs compared to non-infected BMDMs. In contrast, H37Rv:Δ3875 induced significantly less mature IL-1ß production despite eliciting comparable levels of pro-IL-1ß and IL-8 from BMDMs compared to H37Rv and H37Rv:Δ3875C. Blocking either NLRP3 or K+ efflux diminished H37Rv-induced IL-1ß production by BMDMs. Infection of mice intranasally with H37Rv:Δ3875 induced less IL-1ß production in the lungs compared with H37Rv. Intranasal delivery of ESAT-6 but not CFP10 induced production of IL-1ß in mouse lungs and RNA-Seq analysis identified serum amyloid A (SAA) 3 as one of the highly expressed genes in mouse lungs. Infection of mice with H37Rv but not H37Rv:Δ3875 induced expression of lung SAA3 mRNA and protein, consistent with the effect of intranasal delivery of ESAT-6. Silencing SAA3 reduced Mtb-induced IL-1ß production by BMDMs. We conclude that SAA3 plays critical role in ESAT-6 dependent IL-1ß production by macrophages in tuberculosis infection.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Interleucina-1beta/biossíntese , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Proteína Amiloide A Sérica/imunologia , Animais , Feminino , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Amiloide A Sérica/genética , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
4.
PLoS Pathog ; 16(12): e1009132, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33370392

RESUMO

NK cells have been shown to display adaptive traits such as memory formation akin to T and B lymphocytes. Here we show that Zika virus infection induces memory like NK cells that express CD27. Strikingly, these cells exhibit stem-like features that include expansion capacity, self-renewal pathway, differentiation into effector cells, longer telomeres and gene signature associated with hematopoietic stem cell (HSC) progenitors. This subset shared transcriptional and epigenetic changes with memory CD8 T cells, stem cells and stem like T cells. These NK cells with memory and stem cell features, which we term "NK memory stem cells", demonstrated greater antiviral potential than CD27- or naïve CD27+ NK when adoptively transferred to Zika infected mice. Our results also suggest a role for the transcription factor TCF-1 in memory and stemness features of this NK subset. This study defines a unique TCF1hi CD27+ NK subset with memory capacity and stem cell features that play a role in antiviral immunity.


Assuntos
Memória Imunológica/imunologia , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Células-Tronco/imunologia , Infecção por Zika virus/imunologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
5.
Tuberculosis (Edinb) ; 116S: S123-S130, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31103419

RESUMO

Tuberculosis is the most common infectious reason for death and a major cause of pleural effusion globally. To understand the role of chemokines in trafficking of cells during TB pleurisy, we studied the responses to MTB, Ag85A in cells from pleural fluids and peripheral blood. Patients with TB pleural effusions, malignant effusions and asymptomatic healthy controls were enrolled. High expression (p < 0.05) of IP-10, MCP-1, MIG, IL-8, IFN-γ and IL-23 were observed in pleural fluids of TB patients compared to their plasma where expression of RANTES was significantly higher (p < 0.05). On specific stimulation of PFMCs with Ag85A, expression of RANTES was significantly lower in TB compared to NTB patients. We also observed increased expression of T regs and PD1 on CD8+T cells in PFMC of TB patients. Though some of the inflammatory chemokine/cytokines were up-regulated in pleura of TB patients, antigenic stimulation failed to induce them indicating poor antigenic responses at the site. Low expression of RANTES might be a reason for decreased trafficking of cells to the site and dissemination of infection into pleural site. The pattern of RANTES expression in pleural fluid vs serum is interesting. The observations necessitate further studies to investigate the levels of RANTES for its potential biological relevance in TB immunity and its use as a biomarker for diagnosis of pleural TB.


Assuntos
Aciltransferases/imunologia , Antígenos de Bactérias/imunologia , Quimiocina CCL5/metabolismo , Leucócitos Mononucleares/metabolismo , Mycobacterium tuberculosis/imunologia , Derrame Pleural/metabolismo , Tuberculose Pleural/metabolismo , Adulto , Idoso , Biomarcadores/metabolismo , Estudos de Casos e Controles , Quimiocina CCL5/sangue , Quimiotaxia , Regulação para Baixo , Feminino , Interações Hospedeiro-Patógeno , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/microbiologia , Masculino , Pessoa de Meia-Idade , Derrame Pleural/imunologia , Derrame Pleural/microbiologia , Tuberculose Pleural/imunologia , Tuberculose Pleural/microbiologia , Adulto Jovem
6.
J Immunol ; 199(8): 2815-2822, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28855309

RESUMO

In this study, we determined the role of IL-21R signaling in Mycobacterium tuberculosis infection, using IL-21R knockout (KO) mice. A total of 50% of M. tuberculosis H37Rv-infected IL-21R KO mice died in 6 mo compared with no deaths in infected wild type (WT) mice. M. tuberculosis-infected IL-21R KO mice had enhanced bacterial burden and reduced infiltration of Ag-specific T cells in lungs compared with M. tuberculosis-infected WT mice. Ag-specific T cells from the lungs of M. tuberculosis-infected IL-21R KO mice had increased expression of T cell inhibitory receptors, reduced expression of chemokine receptors, proliferated less, and produced less IFN- γ, compared with Ag-specific T cells from the lungs of M. tuberculosis-infected WT mice. T cells from M. tuberculosis-infected IL-21R KO mice were unable to induce optimal macrophage responses to M. tuberculosis. This may be due to a decrease in the Ag-specific T cell population. We also found that IL-21R signaling is associated with reduced expression of a transcriptional factor Eomesodermin and enhanced functional capacity of Ag-specific T cells of M. tuberculosis-infected mice. The sum of our findings suggests that IL-21R signaling is essential for the optimal control of M. tuberculosis infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Receptores de Interleucina-21/metabolismo , Tuberculose/imunologia , Animais , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Interferon gama/metabolismo , Pulmão/microbiologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina-21/genética , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
7.
Tuberculosis (Edinb) ; 101S: S28-S34, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27745787

RESUMO

Abnormalities in hematopoiesis are common in tuberculosis patients and highly prevalent in AIDS patients with tuberculosis coinfection. To explore the potential role of the early secreted antigenic target of 6-kD (ESAT-6) of Mycobacterium tuberculosis (Mtb) in abnormal hematopoiesis in tuberculosis, we studied the effect of ESAT-6 on proliferation and differentiation of in vitro-expanded CD34+ cells isolated from the peripheral blood of the healthy donors. ESAT-6 but not control protein antigen 85A (Ag85A) of Mtb inhibited the proliferation of CD34+ cell derived peripheral blood stem/progenitor cells (PBSPC) in a dose dependent manner when determined by MTT-assay. ESAT-6 but not Ag85A reduced the number of colony forming cells (CFC) of PBSPC by 60-90% as determined by CFC assay by incubation of CD34+ cells in a semi-solid cellulose media in the presence of cytokine cocktail for two weeks. ESAT-6 but not Ag85A increased the percentages of the Annexin-V positive cells and enhanced the cleavage of caspase-3 in PBSPC in a time and dose dependent manner as determined by flow cytometry and Western blot analysis, respectively. ESAT-6 also inhibited murine bone marrow derived non-adherent cell proliferation in response to granulocyte-macrophage colony stimulating factor treatment. We conclude that ESAT-6, an essential virulence factor of Mtb, may contribute to the abnormal hematopoiesis of tuberculosis patients by inhibiting the proliferation and differentiation of hematopoietic cells via apoptosis.


Assuntos
Antígenos de Bactérias/farmacologia , Antígenos CD34/metabolismo , Proteínas de Bactérias/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Mycobacterium tuberculosis/metabolismo , Células-Tronco de Sangue Periférico/efeitos dos fármacos , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Caspase 3/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/imunologia , Células-Tronco de Sangue Periférico/imunologia , Células-Tronco de Sangue Periférico/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
8.
PLoS Pathog ; 12(10): e1005972, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27783671

RESUMO

In this study, we developed a mouse model of type 2 diabetes mellitus (T2DM) using streptozotocin and nicotinamide and identified factors that increase susceptibility of T2DM mice to infection by Mycobacterium tuberculosis (Mtb). All Mtb-infected T2DM mice and 40% of uninfected T2DM mice died within 10 months, whereas all control mice survived. In Mtb-infected mice, T2DM increased the bacterial burden and pro- and anti-inflammatory cytokine and chemokine production in the lungs relative to those in uninfected T2DM mice and infected control mice. Levels of IL-6 also increased. Anti-IL-6 monoclonal antibody treatment of Mtb-infected acute- and chronic-T2DM mice increased survival (to 100%) and reduced pro- and anti-inflammatory cytokine expression. CD11c+ cells were the major source of IL-6 in Mtb-infected T2DM mice. Pulmonary natural killer (NK) cells in Mtb-infected T2DM mice further increased IL-6 production by autologous CD11c+ cells through their activating receptors. Anti-NK1.1 antibody treatment of Mtb-infected acute-T2DM mice increased survival and reduced pro- and anti-inflammatory cytokine expression. Furthermore, IL-6 increased inflammatory cytokine production by T lymphocytes in pulmonary tuberculosis patients with T2DM. Overall, the results suggest that NK-CD11c+ cell interactions increase IL-6 production, which in turn drives the pathological immune response and mortality associated with Mtb infection in diabetic mice.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/imunologia , Células Matadoras Naturais/imunologia , Tuberculose/complicações , Tuberculose/imunologia , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Inflamação/imunologia , Interleucina-6/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Mycobacterium tuberculosis , Reação em Cadeia da Polimerase em Tempo Real , Receptor Cross-Talk/imunologia
9.
Eur J Immunol ; 46(2): 464-79, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26471500

RESUMO

Tissue factor (TF) is a transmembrane glycoprotein that plays an essential role in hemostasis by activating coagulation. TF is also expressed by monocytes/macrophages as part of the innate immune response to infections. In the current study, we determined the role of TF expressed by myeloid cells during Mycobacterium tuberculosis (M. tb) infection by using mice lacking the TF gene in myeloid cells (TF(Δ) ) and human monocyte derived macrophages (MDMs). We found that during M. tb infection, a deficiency of TF in myeloid cells was associated with reduced inducible nitric oxide synthase (iNOS) expression, enhanced arginase 1 (Arg1) expression, enhanced IL-10 production and reduced apoptosis in infected macrophages, which augmented M. tb growth. Our results demonstrate that a deficiency of TF in myeloid cells promotes M2-like phenotype in M .tb infected macrophages. A deficiency in TF expression by myeloid cells was also associated with reduced fibrin deposition and increased matrix metalloproteases (MMP)-2 and MMP-9 mediated inflammation in M. tb infected lungs. Our studies demonstrate that TF expressed by myeloid cells has newly recognized abilities to polarize macrophages and to regulate M. tb growth.


Assuntos
Bacteriemia/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Pneumonia/imunologia , Tromboplastina/metabolismo , Tuberculoma/imunologia , Tuberculose Pulmonar/imunologia , Animais , Bacteriemia/etiologia , Coagulação Sanguínea , Diferenciação Celular , Feminino , Fibrina/genética , Fibrina/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/genética , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/crescimento & desenvolvimento , Pneumonia/etiologia , Tromboplastina/genética , Tuberculoma/etiologia , Tuberculose Pulmonar/complicações
10.
PLoS Pathog ; 11(2): e1004617, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25659138

RESUMO

In this study, we found that a subpopulation of CD4(+)CD25(+) (85% Foxp3(+)) cells from persons with latent tuberculosis infection (LTBI) inhibits growth of M. tuberculosis (M. tb) in human monocyte-derived macrophages (MDMs). A soluble factor, Rho GDP dissociation inhibitor (D4GDI), produced by apoptotic CD4(+)CD25(+) (85% Foxp3(+)) cells is responsible for this inhibition of M. tb growth in human macrophages and in mice. M. tb-expanded CD4(+C)D25(+)Foxp3(+)D4GDI(+) cells do not produce IL-10, TGF-ß and IFN-γ. D4GDI inhibited growth of M. tb in MDMs by enhancing production of IL-1ß, TNF-α and ROS, and by increasing apoptosis of M. tb-infected MDMs. D4GDI was concentrated at the site of disease in tuberculosis patients, with higher levels detected in pleural fluid than in serum. However, in response to M. tb, PBMC from tuberculosis patients produced less D4GDI than PBMC from persons with LTBI. M. tb-expanded CD4+CD25+ (85% Foxp3(+)) cells and D4GDI induced intracellular M. tb to express the dormancy survival regulator DosR and DosR-dependent genes, suggesting that D4GDI induces a non-replicating state in the pathogen. Our study provides the first evidence that a subpopulation of CD4(+)CD25(+) (85% Foxp3+) cells enhances immunity to M. tb, and that production of D4GDI by this subpopulation inhibits M. tb growth.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Tuberculose Latente/imunologia , Macrófagos/microbiologia , Subpopulações de Linfócitos T/imunologia , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico/imunologia , Adolescente , Adulto , Idoso , Animais , Apoptose/imunologia , Separação Celular , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Pessoa de Meia-Idade , Mycobacterium tuberculosis , Reação em Cadeia da Polimerase em Tempo Real , Tuberculose/imunologia , Adulto Jovem
11.
J Infect Dis ; 209(4): 578-87, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24041785

RESUMO

Previously, we found that interleukin 22 (IL-22) inhibits intracellular growth of Mycobacterium tuberculosis in human monocyte-derived macrophages (MDMs). In the current study, we determined the mechanisms underlying these effects. We found that W7, a phagolysosomal fusion inhibitor, abrogates IL-22-dependent M. tuberculosis growth inhibition in MDMs, suggesting that IL-22 acts through enhanced phagolysosomal fusion. Our microarray analysis indicated that recombinant IL-22 (rIL-22) enhances the expression of an intracellular signaling molecule, calgranulin A. This was confirmed by real-time polymerase chain reaction, Western blot, and confocal microscopy. Calgranulin A small interfering RNA (siRNA) abrogated rIL-22-dependent growth inhibition of M. tuberculosis in MDMs. IL-22 enhanced Rab7 expression and downregulated Rab14 expression of M. tuberculosis-infected MDMs, and these effects were reversed by calgranulin A siRNA. These results suggest that M. tuberculosis growth inhibition by IL-22 depends on calgranulin A and enhanced phagolysosomal fusion, which is associated with increased Rab7 and reduced Rab14 expression.


Assuntos
Calgranulina A/biossíntese , Interleucinas/farmacologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/efeitos dos fármacos , Calgranulina A/genética , Calgranulina A/imunologia , Células Cultivadas , Técnicas de Silenciamento de Genes , Humanos , Interleucinas/deficiência , Interleucinas/genética , Interleucinas/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Fagocitose/imunologia , Fagossomos/imunologia , Fagossomos/microbiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Estatísticas não Paramétricas , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7 , Interleucina 22
12.
PLoS One ; 7(9): e45700, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029190

RESUMO

A number of earlier studies reported the occurrence of thrombotic complications, particularly disseminated intravascular coagulation and deep vein thrombosis, in tuberculosis (TB) patients. The aberrant expression of tissue factor (TF), the primary activator of coagulation cascade, is known to be responsible for thrombotic disorders in many diseases including bacterial infections. Further, expression of TF by cells of the monocyte/macrophage lineage is also shown to contribute to the development and progression of local and systemic inflammatory reactions. In the present study, we have investigated whether Mycobacterium tuberculosis (Mtb) infection induces TF expression in macrophages, and various host and pathogenic factors responsible for TF expression. We have tested the effect of live virulent Mtb H37Rv, gamma-irradiated Mtb H37Rv (γ-Mtb) and various components derived from Mtb H37Rv on TF expression in macrophages. The data presented in the manuscript show that both live virulent Mtb and γ-Mtb treatments markedly increased TF activity in macrophages, predominantly in the CD14(+) macrophages. Detailed studies using γ-Mtb showed that the increased TF activity in macrophages following Mtb treatment is the result of TF transcriptional activation. The signaling pathways of TF induction by Mtb appears to be distinct from that of LPS-induced TF expression. Mtb-mediated TF expression is dependent on cooperation of CD14/TLR2/TLR4 and probably yet another unknown receptor/cofactor. Mtb cell wall core components, mycolyl arabinogalactan peptidoglycan (mAGP), phosphatidylinositol mannoside-6 (PIM6) and lipomannan (LM) were identified as factors responsible for induction of TF in the order of mAGP>PIM6>LM. A direct contact between bacteria and macrophage and not Mtb-released soluble factors is critical for TF induction by Mtb. In summary, our data show that Mtb induces TF expression in macrophages and Mtb signaling pathways that elicit TF induction require cooperation of multiple receptors, co-receptors/co-factors including Toll-like receptors. The importance of TF in granuloma formation and containment of Mtb is discussed.


Assuntos
Macrófagos/metabolismo , Mycobacterium tuberculosis/isolamento & purificação , Tromboplastina/metabolismo , Tuberculose/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Receptores de Lipopolissacarídeos/imunologia , Macrófagos/enzimologia , Macrófagos/imunologia , Mycobacterium tuberculosis/patogenicidade , Proteína Quinase C/metabolismo , RNA Mensageiro/genética , Tromboplastina/genética , Ativação Transcricional , Tuberculose/microbiologia , Fator de Necrose Tumoral alfa/metabolismo
13.
J Immunol ; 186(3): 1638-45, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21209279

RESUMO

Macrophages are a major component of the innate immune response, comprising the first line of defense against various intracellular pathogens, including Mycobacterium tuberculosis. In this report, we studied the factors that regulate growth of M. tuberculosis H37Rv in subpopulations of human monocyte-derived macrophages (MDMs). In healthy donors, M. tuberculosis H37Rv grew 5.6-fold more rapidly in CD14(hi) MDMs compared with that in CD14(lo)CD16(+) MDMs. Compared with CD14(lo)CD16(+) cells, M. tuberculosis H37Rv-stimulated CD14(hi) monocytes produced more IL-10 and had increased mRNA expression for c-Maf, a transcription factor that upregulates IL-10 gene expression. c-Maf small interfering RNA (siRNA) inhibited IL-10 production and growth of M. tuberculosis in CD14(hi) cells. Compared with CD14(lo)CD16(+) monocytes, M. tuberculosis H37Rv-stimulated CD14(hi) cells had increased expression of 22 genes whose promoters contained a c-Maf binding site, including hyaluronan synthase 1 (HAS1). c-Maf siRNA inhibited HAS1 expression in M. tuberculosis-stimulated CD14(hi) monocytes, and HAS1 siRNA inhibited growth of M. tuberculosis in CD14(hi) MDMs. M. tuberculosis H37Rv upregulated expression of HAS1 protein and its product, hyaluronan, in CD14(hi) MDMs. We conclude that M. tuberculosis grows more rapidly in CD14(hi) than in CD14(lo)CD16(+) MDMs because CD14(hi) cells have increased expression of c-Maf, which increases production of two key factors (hyaluronan and IL-10) that promote growth of M. tuberculosis.


Assuntos
Receptores de Lipopolissacarídeos/biossíntese , Macrófagos/imunologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/imunologia , Proteínas Proto-Oncogênicas c-maf/fisiologia , Diferenciação Celular/imunologia , Células Cultivadas , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/biossíntese , Humanos , Hialuronan Sintases , Ácido Hialurônico/fisiologia , Interleucina-10/biossíntese , Interleucina-10/fisiologia , Macrófagos/enzimologia , Macrófagos/metabolismo , Monócitos/citologia , Monócitos/enzimologia , Monócitos/imunologia , Mycobacterium tuberculosis/patogenicidade , Proteínas Proto-Oncogênicas c-maf/biossíntese , Virulência/imunologia
14.
J Mol Cell Biol ; 2(5): 237-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20693167

RESUMO

Thymocytes after T-lineage commitment develop in the T-cell pathway. However, in a recent study, Li et al. (2010) demonstrated that inducing to delete Bcl11b gene in these thymocytes, even in mature T cells turns these cells into natural killer (NK) cells during the culture. They called this conversion 'reprogramming', and the reprogrammed killer cells 'ITNK cells'. The ITNK cells possessed tumor-killer ability and did not indiscriminately kill normal cells. This exciting finding represents a major breakthrough towards curing cancer and identifies an important, novel transcription factor in the thymus development.


Assuntos
Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Citotoxicidade Imunológica , Humanos , Imunidade Inata , Imunoterapia , Células Matadoras Naturais/citologia , Camundongos , Neoplasias/genética , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Linfócitos T/citologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
15.
Tuberculosis (Edinb) ; 89 Suppl 1: S77-80, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20006312

RESUMO

Tuberculosis is a leading cause of death from infectious diseases world-wide, and multidrug-resistant (MDR) tuberculosis continues to spread in many parts of the world. MDR tuberculosis is a potential bioterrorist threat, as therapy is prolonged with potentially toxic agents, and the cure rate is much lower than that for treatment of drug-susceptible tuberculosis. Development of methods to enhance innate and adaptive defenses against M. tuberculosis are an attractive means to provide protection against both MDR and drug-susceptible tuberculosis. Before such strategies can be developed, an improved understanding must be gained of the immune response to M. tuberculosis. Our laboratory is mainly focused on understanding the mechanisms by which natural killer (NK) cells lyse M. tuberculosis-infected cells, determining the molecular mechanisms involved in the induction of regulatory T cells (Tregs), and characterizing the mechanisms by which NK cells affect expansion of Tregs in M. tuberculosis infection. As several studies demonstrated defective immune responses in tuberculosis patients, our studies will pinpoint the nature of this defective immune response and permit development of methods to reverse this defect. In the long run, these findings will permit development of novel methods to stimulate immunity against tuberculosis, a strategy that will contribute to development of an effective vaccine to prevent tuberculosis and novel immunotherapy to treat the disease.


Assuntos
Imunidade Adaptativa/imunologia , Antituberculosos/farmacologia , Imunidade Inata , Células Matadoras Naturais/imunologia , Mycobacterium tuberculosis/imunologia , Linfócitos T Reguladores/imunologia , Tuberculose Resistente a Múltiplos Medicamentos/imunologia , Antituberculosos/imunologia , Humanos , Imunoterapia , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Tuberculose Resistente a Múltiplos Medicamentos/terapia
16.
J Immunol ; 183(10): 6639-45, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864591

RESUMO

We determined whether human NK cells could contribute to immune defenses against Mycobacterium tuberculosis through production of IL-22. CD3(-)CD56(+) NK cells produced IL-22 when exposed to autologous monocytes and gamma-irradiated M. tuberculosis, and this depended on the presence of IL-15 and IL-23, but not IL-12 or IL-18. IL-15-stimulated NK cells expressed 10.6 times more DAP10 mRNA compared with control NK cells, and DAP10 siRNA inhibited IL-15-mediated IL-22 production by NK cells. Soluble factors produced by IL-15-activated NK cells inhibited growth of M. tuberculosis in macrophages, and this effect was reversed by anti-IL-22. Addition of rIL-22 to infected macrophages enhanced phagolysosomal fusion and reduced growth of M. tuberculosis. We conclude that NK cells can contribute to immune defenses against M. tuberculosis through production of IL-22, which inhibits intracellular mycobacterial growth by enhancing phagolysosomal fusion. IL-15 and DAP-10 elicit IL-22 production by NK cells in response to M. tuberculosis.


Assuntos
Interleucinas/imunologia , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Fagossomos/imunologia , Tuberculose/imunologia , Humanos , Interleucina-12/farmacologia , Interleucina-15/farmacologia , Interleucina-18/farmacologia , Interleucina-23/farmacologia , Interleucinas/metabolismo , Interleucinas/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/microbiologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/microbiologia , RNA Mensageiro/agonistas , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/imunologia , RNA Interferente Pequeno/metabolismo , Receptores Imunológicos/agonistas , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Proteínas Recombinantes/farmacologia , Tuberculose/microbiologia , Interleucina 22
17.
Transplantation ; 87(12): 1778-86, 2009 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-19543053

RESUMO

BACKGROUND: Immune responses are tempered in immunologically privileged sites including the testis. Previous studies have shown that islet transplantation in the testis significantly prolongs islet allograft survival. However, mechanisms underlying testicular immune privilege and intratesticular allograft survival remain unclear. METHODS: Allogeneic murine islets were transplanted in the testis. Programmed death-1 ligand-1 (PD-L1) expression was detected by immunohistochemstry and real-time polymerase chain reaction. Infiltrating T-cell proliferation was measured by bromodeoxyuridine uptakes, whereas their apoptosis was quantified by terminal deoxynucleotide transferase-mediated dUTP nick-end labeling methods. Transgenic T cells were used to track allospecific memory T-cell generation. RESULTS: We found that programmed death-1 (PD-1):PD-L1 negative costimulation is essential for prolonged survival of intratesticular islet allografts, as blocking PD-L1 or PD-1, but not PD-L2 and cytotoxic T-lymphocyte antigen 4, abrogated long-term survival of intratesticular islet allografts. As controls, blocking PD-1 or PD-L1 did not significantly accelerate the acute rejection of islet allografts transplanted under the renal capsule, a conventional islet-grafting site. We also found for the first time that PD-L1 is constitutively expressed mainly by spermatocytes and spermatids in seminiferous tubules of the testis. Moreover, infiltrating T cells underwent less vigorous proliferation but faster apoptosis in the testis than in the kidney. Blocking PD-1:PD-L1 costimulation largely abolished the suppression of T-cell proliferation and acceleration of T-cell apoptosis. Importantly, testicular immune privilege significantly suppressed the generation and proliferation of donor-specific memory CD8 T cells. CONCLUSIONS: The constitutive expression of PD-L1 in the testis is an important mechanism underlying testicular immune privilege and long-term survival of intratesticular islet allografts.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Antígeno B7-1/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Glicoproteínas de Membrana/imunologia , Peptídeos/imunologia , Testículo/imunologia , Animais , Antígenos de Diferenciação/imunologia , Apoptose , Antígeno B7-H1 , Primers do DNA , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Memória Imunológica , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Morte Celular Programada 1 , RNA Interferente Pequeno/genética , Linfócitos T/imunologia , Transplante Heterotópico/métodos , Transplante Homólogo/imunologia
18.
J Immunol ; 180(3): 1729-36, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18209070

RESUMO

We evaluated the capacity of NK cells to influence expansion of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) in response to microbial Ags, using Mycobacterium tuberculosis as a model. We previously found that Tregs expand when CD4(+) cells and monocytes are exposed to M. tuberculosis. Addition of NK cells that were activated by monokines (IL-12, IL-15, and IL-18) or by exposure to M. tuberculosis-stimulated monocytes reduced Treg expansion in response to M. tuberculosis. NK cell inhibition of Treg expansion was not mediated through IFN-gamma. Activated NK cells lysed expanded, but not freshly isolated Tregs. Although monokines increased NK cell expression of the activating receptors NKp46, NKG2D, 2B4, CD16, and DNAM-1, only anti-NKG2D and anti-NKp46 inhibited NK cell lysis of expanded Tregs. Of five NKG2D ligands, only UL16-binding protein 1 (ULBP1) was up-regulated on M. tuberculosis-expanded Tregs, and anti-ULBP1 inhibited NK cell lysis of expanded Tregs. M. tuberculosis-stimulated monocytes activated NK cells to lyse expanded Tregs, and this was also inhibited by anti-NKG2D and anti-ULBP1, confirming the physiological relevance of this effect. Our study identifies a potential new role for NK cells in maintaining the delicate balance between the regulatory and effector arms of the immune response.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Mycobacterium tuberculosis/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia , Antígenos CD4/análise , Células Cultivadas , Fatores de Transcrição Forkhead/análise , Proteínas Ligadas por GPI , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon gama/metabolismo , Subunidade alfa de Receptor de Interleucina-2/análise , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Monocinas/farmacologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores Imunológicos/agonistas , Receptores de Células Matadoras Naturais
19.
J Immunol ; 172(1): 130-7, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14688318

RESUMO

We studied the role of NK cells in regulating human CD8+ T cell effector function against mononuclear phagocytes infected with the intracellular pathogen Mycobacterium tuberculosis. Depletion of NK cells from PBMC of healthy tuberculin reactors reduced the frequency of M. tuberculosis-responsive CD8+IFN-gamma+ cells and decreased their capacity to lyse M. tuberculosis-infected monocytes. The frequency of CD8+ IFN-gamma+ cells was restored by soluble factors produced by activated NK cells and was dependent on IFN-gamma, IL-15, and IL-18. M. tuberculosis-activated NK cells produced IFN-gamma, activated NK cells stimulated infected monocytes to produce IL-15 and IL-18, and production of IL-15 and IL-18 were inhibited by anti-IFN-gamma. These findings suggest that NK cells maintain the frequency of M. tuberculosis-responsive CD8+IFN-gamma+ T cells by producing IFN-gamma, which elicits secretion of IL-15 and IL-18 by monocytes. These monokines in turn favor expansion of Tc1 CD8+ T cells. The capacity of NK cells to prime CD8+ T cells to lyse M. tuberculosis-infected target cells required cell-cell contact between NK cells and infected monocytes and depended on interactions between the CD40 ligand on NK cells and CD40 on infected monocytes. NK cells link the innate and the adaptive immune responses by optimizing the capacity of CD8+ T cells to produce IFN-gamma and to lyse infected cells, functions that are critical for protective immunity against M. tuberculosis and other intracellular pathogens.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/microbiologia , Citotoxicidade Imunológica , Líquido Intracelular/imunologia , Líquido Intracelular/microbiologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Mycobacterium tuberculosis/imunologia , Anticorpos Bloqueadores/farmacologia , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/imunologia , Ligante de CD40/metabolismo , Ligante de CD40/fisiologia , Antígeno CD56/biossíntese , Antígeno CD56/fisiologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Citotoxicidade Imunológica/imunologia , Humanos , Imunidade Celular , Interferon gama/biossíntese , Interferon gama/fisiologia , Interleucina-15/antagonistas & inibidores , Interleucina-15/biossíntese , Interleucina-18/antagonistas & inibidores , Interleucina-18/biossíntese , Células Matadoras Naturais/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/microbiologia , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Monocinas/fisiologia , Mycobacterium tuberculosis/patogenicidade , Solubilidade , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia , Regulação para Cima/imunologia
20.
AIDS Res Hum Retroviruses ; 19(7): 585-95, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12908936

RESUMO

To characterize the manifestations of coinfection with M. tuberculosis and SIV infection, we studied 12 SIV-infected rhesus monkeys, six of which were infected intrabronchially with a low dose of Mycobacterium tuberculosis H37Rv. In the six coinfected animals, M. tuberculosis antigen-stimulated lung and blood cells produced high concentrations of IFN-gamma but not IL-4 8-16 weeks after infection. Of the three coinfected animals with high levels of plasma viremia, two developed disseminated tuberculosis and the other died of bacterial peritonitis. Of three coinfected animals with moderate levels of plasma viremia, two had no clinical or radiographic evidence of tuberculosis or progressive SIV infection for 6 months after infection. At neuropsy, pulmonary granulomata were observed and acid-fast organisms or M. tuberculosis were present. These clinical, immunologic and pathologic findings are consistent with those in humans with latent tuberculosis infection (LTBI), and suggest that a model of LTBI in SIV-infected primates can be developed. Such a model will permit delineation of the immunologic and microbial factors that characterize LTBI in HIV-infected persons.


Assuntos
Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Tuberculose Pulmonar/complicações , Animais , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Estudos de Viabilidade , Feminino , Regulação da Expressão Gênica , Interferon gama/biossíntese , Interferon gama/genética , Interleucina-4/biossíntese , Interleucina-4/genética , Leucócitos Mononucleares/metabolismo , Pulmão/patologia , Macaca mulatta , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Tuberculoma/patologia , Tuberculose Miliar/complicações , Tuberculose Miliar/imunologia , Tuberculose Miliar/patologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/patologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Carga Viral , Viremia/complicações , Redução de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA