Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Biol ; 22(4): e3002259, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38683873

RESUMO

Antituberculosis drugs, mostly developed over 60 years ago, combined with a poorly effective vaccine, have failed to eradicate tuberculosis. More worryingly, multiresistant strains of Mycobacterium tuberculosis (MTB) are constantly emerging. Innovative strategies are thus urgently needed to improve tuberculosis treatment. Recently, host-directed therapy has emerged as a promising strategy to be used in adjunct with existing or future antibiotics, by improving innate immunity or limiting immunopathology. Here, using high-content imaging, we identified novel 1,2,4-oxadiazole-based compounds, which allow human macrophages to control MTB replication. Genome-wide gene expression analysis revealed that these molecules induced zinc remobilization inside cells, resulting in bacterial zinc intoxication. More importantly, we also demonstrated that, upon treatment with these novel compounds, MTB became even more sensitive to antituberculosis drugs, in vitro and in vivo, in a mouse model of tuberculosis. Manipulation of heavy metal homeostasis holds thus great promise to be exploited to develop host-directed therapeutic interventions.


Assuntos
Antituberculosos , Modelos Animais de Doenças , Macrófagos , Mycobacterium tuberculosis , Oxidiazóis , Tuberculose , Zinco , Animais , Oxidiazóis/farmacologia , Humanos , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Mycobacterium tuberculosis/efeitos dos fármacos , Zinco/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Tuberculose/tratamento farmacológico , Camundongos Endogâmicos C57BL , Feminino , Sinergismo Farmacológico
2.
Elife ; 92020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32369020

RESUMO

Antibiotics are widely used in the treatment of bacterial infections. Although known for their microbicidal activity, antibiotics may also interfere with the host's immune system. Here, we analyzed the effects of bedaquiline (BDQ), an inhibitor of the mycobacterial ATP synthase, on human macrophages. Genome-wide gene expression analysis revealed that BDQ reprogramed cells into potent bactericidal phagocytes. We found that 579 and 1,495 genes were respectively differentially expressed in naive- and M. tuberculosis-infected macrophages incubated with the drug, with an over-representation of lysosome-associated genes. BDQ treatment triggered a variety of antimicrobial defense mechanisms, including phagosome-lysosome fusion, and autophagy. These effects were associated with activation of transcription factor EB, involved in the transcription of lysosomal genes, resulting in enhanced intracellular killing of different bacterial species that were naturally insensitive to BDQ. Thus, BDQ could be used as a host-directed therapy against a wide range of bacterial infections.


The discovery of antibiotic drugs, which treat diseases caused by bacteria, has been a hugely valuable advance in modern medicine. They work by targeting specific cellular processes in bacteria, ultimately stopping them from multiplying or killing them outright. Antibiotics sometimes also affect their human hosts and can cause side-effects, such as gut problems or skin reactions. Recent evidence suggests that antibiotics also have an impact on the human immune system. This may happen either indirectly, by affecting 'friendly' bacteria normally present in the body, or through direct effects on immune cells. In turn, this could change the effectiveness of drug treatments. For example, if an antibiotic weakens immune cells, the body could have difficulty fighting off the existing infection ­ or become more vulnerable to new ones. However, even though new drugs are being introduced to combat the worldwide rise of antibiotic-resistant bacteria, their effects on immunity are still not well understood. For example, bedaquiline is an antibiotic recently developed to treat tuberculosis infections that are resistant to several drugs. Giraud-Gatineau et al. wanted to determine if bedaquiline altered the human immune response to bacterial infection independently from its direct anti-microbial effects. Macrophages engulf foreign particles like bacteria and break them down using enzymes stored within small internal compartments, or 'lysosomes'. Initial experiments using human macrophages, grown both with and without bedaquiline, showed that the drug did not harm the cells and that they grew normally. A combination of microscope imaging and genetic analysis revealed that exposure to bedaquiline not only increased the number of lysosomes within macrophage cells, but also the activity of genes and proteins that increase lysosomes' ability to break down foreign particles. These results suggested that bedaquiline treatment might make macrophages better at fighting infection, even if the drug itself had no direct effect on bacterial cells. Further studies, where macrophages were first treated with bedaquiline and then exposed to different types of bacteria known to be resistant to the drug, confirmed this hypothesis: in every case, the treated macrophages became efficient bacterial killers. In contrast, older anti-tuberculosis drugs did not have any such potentiating effect on the macrophages. This work sheds new light on our how antibiotic drugs can interact with the cells of the human immune system, and can sometimes even boost our innate defences. Such immune-boosting effects could one day be exploited to make more effective treatments against bacterial infections.


Assuntos
Antibacterianos/farmacologia , Diarilquinolinas/farmacologia , Imunidade Inata/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Fagócitos/efeitos dos fármacos , Tuberculose/tratamento farmacológico , Autofagia/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/genética , Lisossomos/metabolismo , Lisossomos/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Fagócitos/imunologia , Fagócitos/metabolismo , Fagócitos/microbiologia , Tuberculose/imunologia , Tuberculose/microbiologia
3.
J Nanobiotechnology ; 17(1): 15, 2019 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-30683129

RESUMO

BACKGROUND: Infectious diseases are still a leading cause of death and, with the emergence of drug resistance, pose a great threat to human health. New drugs and strategies are thus urgently needed to improve treatment efficacy and limit drug-associated side effects. Nanotechnology-based drug delivery systems are promising approaches, offering hope in the fight against drug resistant bacteria. However, how nanocarriers influence the response of innate immune cells to bacterial infection is mostly unknown. RESULTS: Here, we used Mycobacterium tuberculosis as a model of bacterial infection to examine the impact of mannose functionalization of chitosan nanocarriers (CS-NCs) on the human macrophage response. Both ungrafted and grafted CS-NCs were similarly internalized by macrophages, via an actin cytoskeleton-dependent process. Although tri-mannose ligands did not modify the capacity of CS-NCs to escape lysosomal degradation, they profoundly remodeled the response of M. tuberculosis-infected macrophages. mRNA sequencing showed nearly 900 genes to be differentially expressed due to tri-mannose grafting. Unexpectedly, the set of modulated genes was enriched for pathways involved in cell metabolism, particularly oxidative phosphorylation and sugar metabolism. CONCLUSIONS: The ability to modulate cell metabolism by grafting ligands at the surface of nanoparticles may thus be a promising strategy to reprogram immune cells and improve the efficacy of encapsulated drugs.


Assuntos
Infecções Bacterianas/imunologia , Quitosana/química , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Imunidade Inata/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Manose/química , Infecções Bacterianas/microbiologia , Células Cultivadas , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Redes e Vias Metabólicas/efeitos dos fármacos , Mycobacterium tuberculosis/fisiologia , Nanopartículas/química , Nanopartículas/metabolismo , Fagocitose , Transcriptoma/efeitos dos fármacos
4.
Am J Respir Crit Care Med ; 197(6): 801-813, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29161093

RESUMO

RATIONALE: In addition to their well-known function as antibody-producing cells, B lymphocytes can markedly influence the course of infectious or noninfectious diseases via antibody-independent mechanisms. In tuberculosis (TB), B cells accumulate in lungs, yet their functional contribution to the host response remains poorly understood. OBJECTIVES: To document the role of B cells in TB in an unbiased manner. METHODS: We generated the transcriptome of B cells isolated from Mycobacterium tuberculosis (Mtb)-infected mice and validated the identified key pathways using in vitro and in vivo assays. The obtained data were substantiated using B cells from pleural effusion of patients with TB. MEASUREMENTS AND MAIN RESULTS: B cells isolated from Mtb-infected mice displayed a STAT1 (signal transducer and activator of transcription 1)-centered signature, suggesting a role for IFNs in B-cell response to infection. B cells stimulated in vitro with Mtb produced type I IFN, via a mechanism involving the innate sensor STING (stimulator of interferon genes), and antagonized by MyD88 (myeloid differentiation primary response 88) signaling. In vivo, B cells expressed type I IFN in the lungs of Mtb-infected mice and, of clinical relevance, in pleural fluid from patients with TB. Type I IFN expression by B cells induced an altered polarization of macrophages toward a regulatory/antiinflammatory profile in vitro. In vivo, increased provision of type I IFN by B cells in a murine model of B cell-restricted Myd88 deficiency correlated with an enhanced accumulation of regulatory/antiinflammatory macrophages in Mtb-infected lungs. CONCLUSIONS: Type I IFN produced by Mtb-stimulated B cells favors macrophage polarization toward a regulatory/antiinflammatory phenotype during Mtb infection.


Assuntos
Linfócitos B/metabolismo , Interferon Tipo I/metabolismo , Macrófagos/metabolismo , Tuberculose/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis , Transdução de Sinais , Baço/metabolismo , Baço/microbiologia
5.
Artigo em Inglês | MEDLINE | ID: mdl-28824882

RESUMO

Immune responses are essential for the protection of the host against external dangers or infections and are normally efficient in the clearance of invading microbes. However, some intracellular pathogens have developed strategies to replicate and survive within host cells resulting in latent infection associated with strong inflammation. This excessive response can cause cell and tissue damage and lead to the release of the intracellular content, in particular the nucleotide pool, into the extracellular space. Over the last decade, new studies have implicated metabolites from the purinergic pathway in shaping the host immune response against intracellular pathogens and proved their importance in the outcome of the infection. This review aims to summarize how the immune system employs the purinergic system either to fight the pathogen, or to control collateral tissue damage. This will be achieved by focusing on the macrophage response against two intracellular pathogens, the human etiologic agent of tuberculosis, Mycobacterium tuberculosis and the protozoan parasite, Toxoplasma gondii.


Assuntos
Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Transdução de Sinais , Toxoplasma/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Humanos , Imunidade Inata , Macrófagos/microbiologia , Macrófagos/parasitologia , Purinérgicos/metabolismo , Toxoplasmose/imunologia , Tuberculose/imunologia
6.
Sci Rep ; 5: 16882, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26586179

RESUMO

The innate immune system provides the first response to infection and is now recognized to be partially pathogen-specific. Mycobacterium tuberculosis (MTB) is able to subvert the innate immune response and survive inside macrophages. Curiously, only 5-10% of otherwise healthy individuals infected with MTB develop active tuberculosis (TB). We do not yet understand the genetic basis underlying this individual-specific susceptibility. Moreover, we still do not know which properties of the innate immune response are specific to MTB infection. To identify immune responses that are specific to MTB, we infected macrophages with eight different bacteria, including different MTB strains and related mycobacteria, and studied their transcriptional response. We identified a novel subset of genes whose regulation was affected specifically by infection with mycobacteria. This subset includes genes involved in phagosome maturation, superoxide production, response to vitamin D, macrophage chemotaxis, and sialic acid synthesis. We suggest that genetic variants that affect the function or regulation of these genes should be considered candidate loci for explaining TB susceptibility.


Assuntos
Expressão Gênica/imunologia , Imunidade Inata/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Teorema de Bayes , Células Cultivadas , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Predisposição Genética para Doença/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/genética , Macrófagos/metabolismo , Macrófagos/microbiologia , Mycobacterium bovis/imunologia , Mycobacterium bovis/fisiologia , Mycobacterium smegmatis/imunologia , Mycobacterium smegmatis/fisiologia , Mycobacterium tuberculosis/fisiologia , Análise de Componente Principal , Locos de Características Quantitativas/genética , Locos de Características Quantitativas/imunologia , Salmonella typhimurium/imunologia , Salmonella typhimurium/fisiologia , Especificidade da Espécie , Staphylococcus epidermidis/imunologia , Staphylococcus epidermidis/fisiologia , Tuberculose/genética , Tuberculose/imunologia , Tuberculose/microbiologia , Yersinia pseudotuberculosis/imunologia , Yersinia pseudotuberculosis/fisiologia
7.
Infect Immun ; 83(9): 3666-74, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26150535

RESUMO

The immune system needs safeguards that prevent collateral tissue damage mediated by the immune system while enabling an effective response against a pathogen. The purinergic pathway is one such mechanism and finely modulates inflammation by sensing nucleotides in the environment. Extracellular ATP is considered to be a danger signal leading to a proinflammatory response, whereas adenosine is immunosuppressive. CD73, also called ecto-5'-nucleotidase, occupies a strategic position in this pathway, as it is the main enzyme responsible for the generation of adenosine from ATP. Here, we explore the role of CD73 during tuberculosis, a disease characterized by an immune response that is harmful to the host and unable to eradicate Mycobacterium tuberculosis. Using CD73 knockout (KO) mice, we found that CD73 regulates the response to M. tuberculosis infection in vitro and in vivo. Mycobacterium-infected murine macrophages derived from CD73 KO mice secrete more keratinocyte chemoattractant (KC), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6) and release less vascular endothelial growth factor (VEGF) upon ATP stimulation than do those derived from wild-type (WT) mice. In vivo, CD73 limits the early influx of neutrophils to the lungs without affecting bacterial growth and dissemination. Collectively, our results support the view that CD73 fine-tunes antimycobacterial immune responses.


Assuntos
5'-Nucleotidase/imunologia , Infiltração de Neutrófilos/imunologia , Tuberculose Pulmonar/imunologia , 5'-Nucleotidase/deficiência , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/imunologia
8.
J Infect Dis ; 210(5): 824-33, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24604822

RESUMO

Granulomas are the hallmark of Mycobacterium tuberculosis infection. As the host fails to control the bacteria, the center of the granuloma exhibits necrosis resulting from the dying of infected macrophages. The release of the intracellular pool of nucleotides into the surrounding medium may modulate the response of newly infected macrophages, although this has never been investigated. Here, we show that extracellular adenosine triphosphate (ATP) indirectly modulates the expression of 272 genes in human macrophages infected with M. tuberculosis and that it induces their alternative activation. ATP is rapidly hydrolyzed by the ecto-ATPase CD39 into adenosine monophosphate (AMP), and it is AMP that regulates the macrophage response through the adenosine A2A receptor. Our findings reveal a previously unrecognized role for the purinergic pathway in the host response to M. tuberculosis. Dampening inflammation through signaling via the adenosine A2A receptor may limit tissue damage but may also favor bacterial immune escape.


Assuntos
Trifosfato de Adenosina/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Monofosfato de Adenosina/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais
9.
PLoS One ; 7(6): e39080, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22720036

RESUMO

Bacterial infections trigger the expression of type I and II interferon genes but little is known about their effect on type III interferon (IFN-λ) genes, whose products play important roles in epithelial innate immunity against viruses. Here, we studied the expression of IFN-λ genes in cultured human epithelial cells infected with different pathogenic bacteria and in the mouse placenta infected with Listeria monocytogenes. We first showed that in intestinal LoVo cells, induction of IFN-λ genes by L. monocytogenes required bacterial entry and increased further during the bacterial intracellular phase of infection. Other Gram-positive bacteria, Staphylococcus aureus, Staphylococcus epidermidis and Enterococcus faecalis, also induced IFN-λ genes when internalized by LoVo cells. In contrast, Gram-negative bacteria Salmonella enterica serovar Typhimurium, Shigella flexneri and Chlamydia trachomatis did not substantially induce IFN-λ. We also found that IFN-λ genes were up-regulated in A549 lung epithelial cells infected with Mycobacterium tuberculosis and in HepG2 hepatocytes and BeWo trophoblastic cells infected with L. monocytogenes. In a humanized mouse line permissive to fetoplacental listeriosis, IFN-λ2/λ3 mRNA levels were enhanced in placentas infected with L. monocytogenes. In addition, the feto-placental tissue was responsive to IFN-λ2. Together, these results suggest that IFN-λ may be an important modulator of the immune response to Gram-positive intracellular bacteria in epithelial tissues.


Assuntos
Bactérias/patogenicidade , Células Epiteliais/microbiologia , Interferons/genética , Placenta/microbiologia , Animais , Feminino , Camundongos , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Cell Host Microbe ; 10(3): 248-59, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21925112

RESUMO

Mycobacterium tuberculosis thrives within macrophages by residing in phagosomes and preventing them from maturing and fusing with lysosomes. A parallel transcriptional survey of intracellular mycobacteria and their host macrophages revealed signatures of heavy metal poisoning. In particular, mycobacterial genes encoding heavy metal efflux P-type ATPases CtpC, CtpG, and CtpV, and host cell metallothioneins and zinc exporter ZnT1, were induced during infection. Consistent with this pattern of gene modulation, we observed a burst of free zinc inside macrophages, and intraphagosomal zinc accumulation within a few hours postinfection. Zinc exposure led to rapid CtpC induction, and ctpC deficiency caused zinc retention within the mycobacterial cytoplasm, leading to impaired intracellular growth of the bacilli. Thus, the use of P(1)-type ATPases represents a M. tuberculosis strategy to neutralize the toxic effects of zinc in macrophages. We propose that heavy metal toxicity and its counteraction might represent yet another chapter in the host-microbe arms race.


Assuntos
ATPases Bacterianas Próton-Translocadoras/metabolismo , Macrófagos/metabolismo , Mycobacterium tuberculosis/enzimologia , Tuberculose/metabolismo , Zinco/metabolismo , Animais , ATPases Bacterianas Próton-Translocadoras/genética , Células Cultivadas , Feminino , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Tuberculose/microbiologia , Zinco/toxicidade
11.
J Exp Med ; 206(10): 2205-20, 2009 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-19770268

RESUMO

The C-type lectin dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN) mediates the innate immune recognition of microbial carbohydrates. We investigated the function of this molecule in the host response to pathogens in vivo, by generating mouse lines lacking the DC-SIGN homologues SIGNR1, SIGNR3, and SIGNR5. Resistance to Mycobacterium tuberculosis was impaired only in SIGNR3-deficient animals. SIGNR3 was expressed in lung phagocytes during infection, and interacted with M. tuberculosis bacilli and mycobacterial surface glycoconjugates to induce secretion of critical host defense inflammatory cytokines, including tumor necrosis factor (TNF). SIGNR3 signaling was dependent on an intracellular tyrosine-based motif and the tyrosine kinase Syk. Thus, the mouse DC-SIGN homologue SIGNR3 makes a unique contribution to protection of the host against a pulmonary bacterial pathogen.


Assuntos
Moléculas de Adesão Celular/fisiologia , Lectinas Tipo C/fisiologia , Receptores de Superfície Celular/fisiologia , Tuberculose/imunologia , Animais , Antígenos CD/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Glicoconjugados/metabolismo , Interleucina-6/biossíntese , Lipopolissacarídeos/metabolismo , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Transdução de Sinais , Receptor 2 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/biossíntese
12.
PLoS One ; 3(1): e1403, 2008 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18167562

RESUMO

BACKGROUND: Transcriptional profiling using microarrays provides a unique opportunity to decipher host pathogen cross-talk on the global level. Here, for the first time, we have been able to investigate gene expression changes in both Mycobacterium tuberculosis, a major human pathogen, and its human host cells, macrophages and dendritic cells. METHODOLOGY/PRINCIPAL FINDINGS: In addition to common responses, we could identify eukaryotic and microbial transcriptional signatures that are specific to the cell type involved in the infection process. In particular M. tuberculosis shows a marked stress response when inside dendritic cells, which is in accordance with the low permissivity of these specialized phagocytes to the tubercle bacillus and to other pathogens. In contrast, the mycobacterial transcriptome inside macrophages reflects that of replicating bacteria. On the host cell side, differential responses to infection in macrophages and dendritic cells were identified in genes involved in oxidative stress, intracellular vesicle trafficking and phagosome acidification. CONCLUSIONS/SIGNIFICANCE: This study provides the proof of principle that probing the host and the microbe transcriptomes simultaneously is a valuable means to accessing unique information on host pathogen interactions. Our results also underline the extraordinary plasticity of host cell and pathogen responses to infection, and provide a solid framework to further understand the complex mechanisms involved in immunity to M. tuberculosis and in mycobacterial adaptation to different intracellular environments.


Assuntos
Células Dendríticas/microbiologia , Perfilação da Expressão Gênica , Macrófagos/microbiologia , Mycobacterium tuberculosis/genética , Transcrição Gênica , Células Dendríticas/imunologia , Humanos , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/fisiologia
13.
Infect Immun ; 75(1): 504-7, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17030567

RESUMO

Using signature-tagged transposon mutagenesis, we isolated 23 Mycobacterium tuberculosis mutants, corresponding to 21 genes or genetic regions, attenuated in their ability to parasitize human macrophages. Mutants disrupted in the ABC transporter-encoding genes Rv0986 and Rv0987 were further characterized as being impaired in their ability to bind to host cells.


Assuntos
Elementos de DNA Transponíveis/genética , Genes Bacterianos , Macrófagos/parasitologia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Animais , Proteínas de Bactérias/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese
14.
PLoS One ; 1: e43, 2006 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-17183672

RESUMO

BACKGROUND: Mycobacterium tuberculosis, the etiological agent of tuberculosis (TB), has the ability to persist in its human host for exceptionally long periods of time. However, little is known about the location of the bacilli in latently infected individuals. Long-term mycobacterial persistence in the lungs has been reported, but this may not sufficiently account for strictly extra-pulmonary TB, which represents 10-15% of the reactivation cases. METHODOLOGY/PRINCIPAL FINDINGS: We applied in situ and conventional PCR to sections of adipose tissue samples of various anatomical origins from 19 individuals from Mexico and 20 from France who had died from causes other than TB. M. tuberculosis DNA could be detected by either or both techniques in fat tissue surrounding the kidneys, the stomach, the lymph nodes, the heart and the skin in 9/57 Mexican samples (6/19 individuals), and in 8/26 French samples (6/20 individuals). In addition, mycobacteria could be immuno-detected in perinodal adipose tissue of 1 out of 3 biopsy samples from individuals with active TB. In vitro, using a combination of adipose cell models, including the widely used murine adipose cell line 3T3-L1, as well as primary human adipocytes, we show that after binding to scavenger receptors, M. tuberculosis can enter within adipocytes, where it accumulates intracytoplasmic lipid inclusions and survives in a non-replicating state that is insensitive to the major anti-mycobacterial drug isoniazid. CONCLUSIONS/SIGNIFICANCE: Given the abundance and the wide distribution of the adipose tissue throughout the body, our results suggest that this tissue, among others, might constitute a vast reservoir where the tubercle bacillus could persist for long periods of time, and avoid both killing by antimicrobials and recognition by the host immune system. In addition, M. tuberculosis-infected adipocytes might provide a new model to investigate dormancy and to evaluate new drugs for the treatment of persistent infection.


Assuntos
Tecido Adiposo/microbiologia , Tuberculose Latente/microbiologia , Mycobacterium tuberculosis/patogenicidade , Células 3T3-L1 , Adipócitos/microbiologia , Adipócitos/ultraestrutura , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA/genética , DNA Bacteriano/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Tuberculose Latente/imunologia , Camundongos , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/isolamento & purificação , Receptores Depuradores/fisiologia , Tuberculose/microbiologia
15.
PLoS Med ; 2(12): e381, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16279841

RESUMO

BACKGROUND: Interplays between Mycobacterium tuberculosis, the etiological agent of tuberculosis (TB) in human and host professional phagocytes, namely macrophages (Mphis) and dendritic cells (DCs), are central to immune protection against TB and to TB pathogenesis. We and others have recently shown that the C-type lectin dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN; CD209) mediates important interactions between mycobacteria and human monocyte-derived DCs (MoDCs) in vitro. METHODS AND FINDINGS: In order to explore the possible role of DC-SIGN in M. tuberculosis infection in vivo, we have analysed DC-SIGN expression in broncho-alveolar lavage (BAL) cells from patients with TB (n = 40) or with other non-mycobacterial lung pathologies, namely asthma (n = 14) and sarcoidosis (n = 11), as well as from control individuals (n = 9). We show that in patients with TB, up to 70% of alveolar Mphis express DC-SIGN. By contrast, the lectin is barely detected in alveolar Mphis from all other individuals. Flow cytometry, RT-PCR, and enzyme-linked immunosorbent assay analyses of BAL-derived fluids and cells indicated that M. tuberculosis infection induces DC-SIGN expression in alveolar Mphis by a mechanism that is independent of Toll-like receptor-4, interleukin (IL)-4, and IL-13. This mechanism most likely relies on the secretion of soluble host and/or mycobacterial factors that have yet to be identified, as both infected and uninfected bystander Mphis were found to express DC-SIGN in the presence of M. tuberculosis. Immunohistochemical examination of lung biopsy samples from patients with TB showed that the bacilli concentrate in pulmonary regions enriched in DC-SIGN-expressing alveolar Mphis in vivo. Ex vivo binding and inhibition of binding experiments further revealed that DC-SIGN-expressing alveolar Mphis constitute preferential target cells for M. tuberculosis, as compared to their DC-SIGN- counterparts. In contrast with what has been reported previously in MoDCs in vitro, ex vivo DC-SIGN ligation by mycobacterial products failed to induce IL-10 secretion by alveolar Mphis, and IL-10 was not detected in BALs from patients with TB. CONCLUSION: Altogether, our results provide further evidence for an important role of DC-SIGN during TB in humans. DC-SIGN induction in alveolar Mphis may have important consequences on lung colonization by the tubercle bacillus, and on pulmonary inflammatory and immune responses in the infected host.


Assuntos
Moléculas de Adesão Celular/fisiologia , Lectinas Tipo C/fisiologia , Receptores de Superfície Celular/fisiologia , Tuberculose Pulmonar/fisiopatologia , Estudos de Casos e Controles , Moléculas de Adesão Celular/biossíntese , Humanos , Lectinas Tipo C/biossíntese , Macrófagos Alveolares , Mycobacterium tuberculosis/patogenicidade , Receptores de Superfície Celular/biossíntese
16.
Trends Microbiol ; 11(6): 259-63, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12823942

RESUMO

Mammals have evolved surface pattern recognition receptors, such as the Toll-like receptors, to initiate defenses against pathogens, including mycobacterium. In turn, microbes have developed strategies to circumvent defenses of their host and establish persistent infections. Mycobacterium tuberculosis, one of the most successful pathogens worldwide, has the ability to parasitize and manipulate phagocytic cells of its human host. A set of recent reports has shed light on exploitation of phagocyte surface lectins by the tubercle bacillus. These findings could lead the way to innovative therapeutic approaches.


Assuntos
Lectinas/metabolismo , Mycobacterium tuberculosis/patogenicidade , Fagócitos/microbiologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/microbiologia , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Modelos Biológicos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Fagócitos/metabolismo , Receptores de Superfície Celular/metabolismo
17.
J Immunol ; 170(4): 1939-48, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12574362

RESUMO

Dendritic cells (DCs) are likely to play a key role in immunity against Mycobacterium tuberculosis, but the fate of the bacterium in these cells is still unknown. Here we report that, unlike macrophages (Mphis), human monocyte-derived DCs are not permissive for the growth of virulent M. tuberculosis H37Rv. Mycobacterial vacuoles are neither acidic nor fused with host cell lysosomes in DCs, in a mode similar to that seen in mycobacterial infection of Mphis. However, uptake of the fluid phase marker dextran, and of transferrin, as well as accumulation of the recycling endosome-specific small GTPase Rab11 onto the mycobacterial phagosome, are almost abolished in infected DCs, but not in Mphis. Moreover, communication between mycobacterial phagosomes and the host-cell biosynthetic pathway is impaired, given that <10% of M. tuberculosis vacuoles in DCs stained for the endoplasmic reticulum-specific proteins Grp78/BiP and calnexin. This correlates with the absence of the fusion factor N-ethylmaleimide-sensitive factor onto the vacuolar membrane in this cell type. Trafficking between the vacuoles and the host cell recycling and biosynthetic pathways is strikingly reduced in DCs, which is likely to impair access of intracellular mycobacteria to essential nutrients and may thus explain the absence of mycobacterial growth in this cell type. This unique location of M. tuberculosis in DCs is compatible with their T lymphocyte-stimulating functions, because M. tuberculosis-infected DCs have the ability to specifically induce cytokine production by autologous T lymphocytes from presensitized individuals. DCs have evolved unique subcellular trafficking mechanisms to achieve their Ag-presenting functions when infected by intracellular mycobacteria.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Líquido Intracelular/imunologia , Líquido Intracelular/microbiologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/imunologia , Apresentação de Antígeno , Diferenciação Celular/imunologia , Divisão Celular/imunologia , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Chaperona BiP do Retículo Endoplasmático , Humanos , Imunidade Inata , Líquido Intracelular/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Fagossomos/imunologia , Fagossomos/microbiologia , Biossíntese de Proteínas , Especificidade da Espécie , Tuberculina/imunologia , Tuberculina/metabolismo , Vacúolos/imunologia , Vacúolos/microbiologia
18.
J Leukoc Biol ; 72(6): 1180-9, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12488500

RESUMO

That monocytes can differentiate into macrophages or dendritic cells (DCs) makes them an essential link between innate and adaptive immunity. However, little is known about how interactions with pathogens or T cells influence monocyte engagement toward DCs. We approached this point in cultures where granulocyte macrophage-colony stimulating factor (GM-CSF) and interleukin (IL)-4 induced monocytes to differentiate into immature DCs. Activating monocytes with soluble CD40 ligand (CD40L) led to accelerated differentiation toward mature CD83(+) DCs with up-regulated human leukocyte antigen-DR, costimulatory molecules and CD116 (GM-CSF receptor), and down-regulation of molecules involved in antigen capture. Monocytes primed by phagocytosis of antibody-opsonized, killed Escherichia coli differentiated into DCs with an immature phenotype, whereas Zymosan priming yielded active DCs with an intermediate phenotype. Accordingly, DCs obtained from cultures with CD40L or after Zymosan priming had a decreased capacity to endocytose dextran, but only DCs cultured with CD40L had increased capacity to stimulate allogeneic T cells. DCs obtained after E. coli or Zymosan priming of monocytes produced high levels of proinflammatory tumor necrosis factor alpha and IL-6 as well as of regulatory IL-10, but they produced IL-12p70 only after secondary CD40 ligation. Thus, CD40 ligation on monocytes accelerates the maturation of DCs in the presence of GM-CSF/IL-4, whereas phagocytosis of different microorganisms does not alter and even facilitates their potential to differentiate into immature or active DCs, the maturation of which can be completed upon CD40 ligation. In vivo, such differences may correspond to DCs with different trafficking and T helper cell-stimulating capacities that could differently affect induction of adaptive immune responses to infections.


Assuntos
Ligante de CD40/farmacologia , Células Dendríticas/citologia , Monócitos/citologia , Fagocitose/fisiologia , Antígenos CD/análise , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos , Imunofenotipagem , Teste de Cultura Mista de Linfócitos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Ligação Proteica , Receptores de Quimiocinas/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA