Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
ACS Chem Biol ; 19(3): 707-717, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-38442242

RESUMO

Surface lipids on pathogenic mycobacteria modulate infection outcomes by regulating host immune responses. Phenolic glycolipid (PGL) is a host-modulating surface lipid that varies among clinical Mycobacterium tuberculosis strains. PGL is also found in Mycobacterium marinum, where it promotes infection of zebrafish through effects on the innate immune system. Given the important role this lipid plays in the host-pathogen relationship, tools for profiling its abundance, spatial distribution, and dynamics are needed. Here, we report a strategy for imaging PGL in live mycobacteria using bioorthogonal metabolic labeling. We functionalized the PGL precursor p-hydroxybenzoic acid (pHB) with an azide group (3-azido pHB). When fed to mycobacteria, 3-azido pHB was incorporated into the cell surface, which could then be visualized via the bioorthogonal conjugation of a fluorescent probe. We confirmed that 3-azido pHB incorporates into PGL using mass spectrometry methods and demonstrated selectivity for PGL-producing M. marinum and M. tuberculosis strains. Finally, we applied this metabolic labeling strategy to study the dynamics of PGL within the mycobacterial membrane. This new tool enables visualization of PGL that may facilitate studies of mycobacterial pathogenesis.


Assuntos
Mycobacterium marinum , Mycobacterium tuberculosis , Animais , Glicolipídeos/metabolismo , Fatores de Virulência/metabolismo , Peixe-Zebra , Mycobacterium tuberculosis/metabolismo , Mycobacterium marinum/metabolismo
2.
JACS Au ; 1(9): 1368-1379, 2021 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-34604847

RESUMO

There is an urgent need for point-of-care tuberculosis (TB) diagnostic methods that are fast, inexpensive, and operationally simple. Here, we report on a bright solvatochromic dye trehalose conjugate that specifically detects Mycobacterium tuberculosis (Mtb) in minutes. 3-Hydroxychromone (3HC) dyes, known for having high fluorescence quantum yields, exhibit shifts in fluorescence intensity in response to changes in environmental polarity. We synthesized two analogs of 3HC-trehalose conjugates (3HC-2-Tre and 3HC-3-Tre) and determined that 3HC-3-Tre has exceptionally favorable properties for Mtb detection. 3HC-3-Tre-labeled mycobacterial cells displayed a 10-fold increase in fluorescence intensity compared to our previous reports on the dye 4,4-N,N-dimethylaminonapthalimide (DMN-Tre). Excitingly, we detected fluorescent Mtb cells within 10 min of probe treatment. Thus, 3HC-3-Tre permits rapid visualization of mycobacteria that ultimately could empower improved Mtb detection at the point-of-care in low-resource settings.

3.
Elife ; 92020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33226343

RESUMO

Several virulence lipids populate the outer cell wall of pathogenic mycobacteria. Phthiocerol dimycocerosate (PDIM), one of the most abundant outer membrane lipids, plays important roles in both defending against host antimicrobial programs and in evading these programs altogether. Immediately following infection, mycobacteria rely on PDIM to evade Myd88-dependent recruitment of microbicidal monocytes which can clear infection. To circumvent the limitations in using genetics to understand virulence lipids, we developed a chemical approach to track PDIM during Mycobacterium marinum infection of zebrafish. We found that PDIM's methyl-branched lipid tails enabled it to spread into host epithelial membranes to prevent immune activation. Additionally, PDIM's affinity for cholesterol promoted this phenotype; treatment of zebrafish with statins, cholesterol synthesis inhibitors, decreased spreading and provided protection from infection. This work establishes that interactions between host and pathogen lipids influence mycobacterial infectivity and suggests the use of statins as tuberculosis preventive therapy by inhibiting PDIM spread.


Assuntos
Membrana Celular/microbiologia , Células Epiteliais/microbiologia , Lipídeos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/patogenicidade , Fatores de Virulência/metabolismo , Células A549 , Animais , Animais Geneticamente Modificados , Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lipídeos/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/microbiologia , Estrutura Molecular , Infecções por Mycobacterium não Tuberculosas/metabolismo , Infecções por Mycobacterium não Tuberculosas/prevenção & controle , Mycobacterium marinum/efeitos dos fármacos , Mycobacterium marinum/genética , Mycobacterium marinum/metabolismo , Relação Estrutura-Atividade , Células THP-1 , Virulência , Fatores de Virulência/química , Peixe-Zebra
4.
Cell Host Microbe ; 24(4): 514-525.e6, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30308157

RESUMO

Mycobacterial infection leads to the formation of characteristic immune aggregates called granulomas, a process accompanied by dramatic remodeling of the host vasculature. As granuloma angiogenesis favors the infecting mycobacteria, it may be actively promoted by bacterial determinants during infection. Using Mycobacterium marinum-infected zebrafish as a model, we identify the enzyme proximal cyclopropane synthase of alpha-mycolates (PcaA) as an important bacterial determinant of granuloma-associated angiogenesis. cis-Cyclopropanation of mycobacterial mycolic acids by pcaA drives the activation of host Vegf signaling within granuloma macrophages. Cyclopropanation of the mycobacterial cell wall glycolipid trehalose dimycolate is both required and sufficient to induce robust host angiogenesis. Inducible genetic inhibition of angiogenesis and Vegf signaling during granuloma formation results in bacterial growth deficits. Together, these data reveal a mechanism by which PcaA-mediated cis-cyclopropanation of mycolic acids promotes bacterial growth and dissemination in vivo by eliciting granuloma vascularization and suggest potential approaches for host-directed therapies.


Assuntos
Proteínas de Bactérias/metabolismo , Metiltransferases/metabolismo , Mycobacterium marinum/enzimologia , Neovascularização Patológica/microbiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Tuberculoma/microbiologia , Inibidores da Angiogênese/farmacologia , Animais , Proteínas de Bactérias/genética , Fatores Corda/metabolismo , Modelos Animais de Doenças , Humanos , Indazóis , Macrófagos/imunologia , Macrófagos/microbiologia , Metiltransferases/genética , Infecções por Mycobacterium não Tuberculosas/imunologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidade , Ácidos Micólicos/metabolismo , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Pirimidinas/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Transdução de Sinais , Sulfonamidas/farmacologia , Tuberculoma/imunologia , Tuberculoma/patologia , Peixe-Zebra
5.
Cell ; 170(5): 973-985.e10, 2017 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-28841420

RESUMO

Mycobacterium leprae causes leprosy and is unique among mycobacterial diseases in producing peripheral neuropathy. This debilitating morbidity is attributed to axon demyelination resulting from direct interaction of the M. leprae-specific phenolic glycolipid 1 (PGL-1) with myelinating glia and their subsequent infection. Here, we use transparent zebrafish larvae to visualize the earliest events of M. leprae-induced nerve damage. We find that demyelination and axonal damage are not directly initiated by M. leprae but by infected macrophages that patrol axons; demyelination occurs in areas of intimate contact. PGL-1 confers this neurotoxic response on macrophages: macrophages infected with M. marinum-expressing PGL-1 also damage axons. PGL-1 induces nitric oxide synthase in infected macrophages, and the resultant increase in reactive nitrogen species damages axons by injuring their mitochondria and inducing demyelination. Our findings implicate the response of innate macrophages to M. leprae PGL-1 in initiating nerve damage in leprosy.


Assuntos
Antígenos de Bactérias/metabolismo , Modelos Animais de Doenças , Glicolipídeos/metabolismo , Hanseníase/microbiologia , Hanseníase/patologia , Macrófagos/imunologia , Mycobacterium leprae/fisiologia , Animais , Axônios/metabolismo , Axônios/patologia , Doenças Desmielinizantes , Larva/crescimento & desenvolvimento , Hanseníase/imunologia , Mycobacterium marinum/metabolismo , Bainha de Mielina/química , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Neuroglia/metabolismo , Neuroglia/patologia , Óxido Nítrico/metabolismo , Peixe-Zebra
6.
Immunity ; 47(3): 552-565.e4, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28844797

RESUMO

Mycobacterium tuberculosis (Mtb) enters the host in aerosol droplets deposited in lung alveoli, where the bacteria first encounter lung-resident alveolar macrophages. We studied the earliest mycobacterium-macrophage interactions in the optically transparent zebrafish. First-responding resident macrophages phagocytosed and eradicated infecting mycobacteria, suggesting that to establish a successful infection, mycobacteria must escape out of the initially infected resident macrophage into growth-permissive monocytes. We defined a critical role for mycobacterial membrane phenolic glycolipid (PGL) in engineering this transition. PGL activated the STING cytosolic sensing pathway in resident macrophages, inducing the production of the chemokine CCL2, which in turn recruited circulating CCR2+ monocytes toward infection. Transient fusion of infected macrophages with CCR2+ monocytes enabled bacterial transfer and subsequent dissemination, and interrupting this transfer so as to prolong mycobacterial sojourn in resident macrophages promoted clearing of infection. Human alveolar macrophages produced CCL2 in a PGL-dependent fashion following infection, arguing for the potential of PGL-blocking interventions or PGL-targeting vaccine strategies in the prevention of tuberculosis. VIDEO ABSTRACT.


Assuntos
Glicolipídeos/imunologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Mycobacterium tuberculosis/imunologia , Animais , Quimiocina CCL2/metabolismo , Quimiotaxia/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/fisiologia , Proteínas de Membrana/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Mutação , Mycobacterium tuberculosis/genética , Especificidade de Órgãos/imunologia , Tuberculose/imunologia , Tuberculose/metabolismo , Tuberculose/microbiologia , Peixe-Zebra
7.
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
8.
J Exp Med ; 212(5): 715-28, 2015 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-25918344

RESUMO

Immune control of persistent infection with Mycobacterium tuberculosis (Mtb) requires a sustained pathogen-specific CD4 T cell response; however, the molecular pathways governing the generation and maintenance of Mtb protective CD4 T cells are poorly understood. Using MHCII tetramers, we show that Mtb-specific CD4 T cells are subject to ongoing antigenic stimulation. Despite this chronic stimulation, a subset of PD-1(+) cells is maintained within the lung parenchyma during tuberculosis (TB). When transferred into uninfected animals, these cells persist, mount a robust recall response, and provide superior protection to Mtb rechallenge when compared to terminally differentiated Th1 cells that reside preferentially in the lung-associated vasculature. The PD-1(+) cells share features with memory CD4 T cells in that their generation and maintenance requires intrinsic Bcl6 and intrinsic ICOS expression. Thus, the molecular pathways required to maintain Mtb-specific CD4 T cells during ongoing infection are similar to those that maintain memory CD4 T cells in scenarios of antigen deprivation. These results suggest that vaccination strategies targeting the ICOS and Bcl6 pathways in CD4 T cells may provide new avenues to prevent TB.


Assuntos
Proteínas de Ligação a DNA/imunologia , Memória Imunológica , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Mycobacterium tuberculosis/imunologia , Células Th1/imunologia , Tuberculose Pulmonar/imunologia , Animais , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/imunologia , Imunidade Celular/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-6 , Células Th1/patologia , Tuberculose Pulmonar/genética , Tuberculose Pulmonar/patologia
9.
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
10.
Nature ; 505(7482): 218-22, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24336213

RESUMO

The evolutionary survival of Mycobacterium tuberculosis, the cause of human tuberculosis, depends on its ability to invade the host, replicate, and transmit infection. At its initial peripheral infection site in the distal lung airways, M. tuberculosis infects macrophages, which transport it to deeper tissues. How mycobacteria survive in these broadly microbicidal cells is an important question. Here we show in mice and zebrafish that M. tuberculosis, and its close pathogenic relative Mycobacterium marinum, preferentially recruit and infect permissive macrophages while evading microbicidal ones. This immune evasion is accomplished by using cell-surface-associated phthiocerol dimycoceroserate (PDIM) lipids to mask underlying pathogen-associated molecular patterns (PAMPs). In the absence of PDIM, these PAMPs signal a Toll-like receptor (TLR)-dependent recruitment of macrophages that produce microbicidal reactive nitrogen species. Concordantly, the related phenolic glycolipids (PGLs) promote the recruitment of permissive macrophages through a host chemokine receptor 2 (CCR2)-mediated pathway. Thus, we have identified coordinated roles for PDIM, known to be essential for mycobacterial virulence, and PGL, which (along with CCR2) is known to be associated with human tuberculosis. Our findings also suggest an explanation for the longstanding observation that M. tuberculosis initiates infection in the relatively sterile environment of the lower respiratory tract, rather than in the upper respiratory tract, where resident microflora and inhaled environmental microbes may continually recruit microbicidal macrophages through TLR-dependent signalling.


Assuntos
Evasão da Resposta Imune , Macrófagos/microbiologia , Lipídeos de Membrana/metabolismo , Mycobacterium/fisiologia , Animais , Feminino , Glicolipídeos/imunologia , Glicolipídeos/metabolismo , Lipídeos/biossíntese , Lipídeos/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Mycobacterium tuberculosis/fisiologia , Receptores CCR2/metabolismo , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Virulência/imunologia , Peixe-Zebra/microbiologia
11.
Cell Host Microbe ; 12(3): 301-12, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22980327

RESUMO

Neutrophils are typically the first responders in host defense against invading pathogens, which they destroy by both oxidative and nonoxidative mechanisms. However, despite a longstanding recognition of neutrophil presence at disease sites in tuberculosis, their role in defense against mycobacteria is unclear. Here we exploit the genetic tractability and optical transparency of zebrafish to monitor neutrophil behavior and its consequences during infection with Mycobacterium marinum, a natural fish pathogen. In contrast to macrophages, neutrophils do not interact with mycobacteria at initial infection sites. Neutrophils are subsequently recruited to the nascent granuloma in response to signals from dying infected macrophages within the granuloma, which they phagocytose. Some neutrophils then rapidly kill the internalized mycobacteria through NADPH oxidase-dependent mechanisms. Our results provide a mechanistic link to the observed patterns of neutrophils in human tuberculous granulomas and the susceptibility of humans with chronic granulomatous disease to mycobacterial infection.


Assuntos
Granuloma/imunologia , Granuloma/microbiologia , Viabilidade Microbiana/efeitos dos fármacos , Mycobacterium marinum/imunologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Estresse Oxidativo , Animais , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Dados de Sequência Molecular , Mycobacterium marinum/efeitos dos fármacos , NADP/metabolismo , Oxirredução , Oxirredutases/metabolismo , Análise de Sequência de DNA , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia
12.
J Immunol ; 184(11): 6522-8, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20421649

RESUMO

DAP12 is an adapter protein that associates with several receptors in macrophages. Little is known about the biological role of DAP12 in alveolar macrophages. In genome-wide profiling, we previously found that two DAP12-associated receptors, myeloid DAP12-associated lectin-1 and triggering receptor expressed on myeloid cells 2 (TREM2), were highly induced in alveolar macrophages from habitual smokers. Here, we found that transcript levels for these receptors in alveolar macrophages increased with packs per day of cigarettes smoked and expression of TREM2 protein was increased in lung macrophages of former smokers with emphysema compared with that in controls. In vitro, cigarette smoke directly induced expression of myeloid DAP12-associated lectin-1 and TREM2 and activation of DAP12 signaling in mouse macrophages. To determine whether DAP12 plays a role in cigarette smoke-induced pulmonary inflammation, we exposed wild-type and DAP12-deficient mice to chronic cigarette smoke and found significant reduction in recruitment of alveolar macrophages in DAP12-deficient mice. Because cigarette smoking induces the macrophage chemoattractant CCL2, we tested the chemotactic ability of DAP12-deficient macrophages and found abrogation of chemotaxis toward CCL2 in vitro. Airway administration of CCL2 also resulted in a significant reduction of macrophage recruitment to the lungs of DAP12-deficient mice compared with that in controls. DAP12 was also required for normal macrophage migration in a "scratch" assay. Reconstitution studies revealed that phosphorylation of the DAP12 ITAM was required for normal migration in vitro and association with TREM2 was sufficient for normal migration. These findings indicate that DAP12, possibly through association with TREM2, contributes to alveolar macrophage chemotaxis and recruitment to the lung and may mediate macrophage accumulation in lung diseases such as emphysema.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quimiocina CCL2/metabolismo , Quimiotaxia de Leucócito/imunologia , Pulmão/imunologia , Macrófagos Alveolares/metabolismo , Proteínas de Membrana/metabolismo , Fumaça/efeitos adversos , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Western Blotting , Separação Celular , Quimiocina CCL2/imunologia , Citometria de Fluxo , Imunofluorescência , Humanos , Imunoprecipitação , Macrófagos Alveolares/imunologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doença Pulmonar Obstrutiva Crônica/imunologia , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...