Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(20): 3720-3738.e13, 2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-36103894

RESUMO

Necrosis of macrophages in the granuloma, the hallmark immunological structure of tuberculosis, is a major pathogenic event that increases host susceptibility. Through a zebrafish forward genetic screen, we identified the mTOR kinase, a master regulator of metabolism, as an early host resistance factor in tuberculosis. We found that mTOR complex 1 protects macrophages from mycobacterium-induced death by enabling infection-induced increases in mitochondrial energy metabolism fueled by glycolysis. These metabolic adaptations are required to prevent mitochondrial damage and death caused by the secreted mycobacterial virulence determinant ESAT-6. Thus, the host can effectively counter this early critical mycobacterial virulence mechanism simply by regulating energy metabolism, thereby allowing pathogen-specific immune mechanisms time to develop. Our findings may explain why Mycobacterium tuberculosis, albeit humanity's most lethal pathogen, is successful in only a minority of infected individuals.


Assuntos
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculose , Animais , Mycobacterium tuberculosis/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Peixe-Zebra
2.
Cell ; 185(24): 4507-4525.e18, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36356582

RESUMO

The human pathogen Mycobacterium tuberculosis typically causes lung disease but can also disseminate to other tissues. We identified a M. tuberculosis (Mtb) outbreak presenting with unusually high rates of extrapulmonary dissemination and bone disease. We found that the causal strain carried an ancestral full-length version of the type VII-secreted effector EsxM rather than the truncated version present in other modern Mtb lineages. The ancestral EsxM variant exacerbated dissemination through enhancement of macrophage motility, increased egress of macrophages from established granulomas, and alterations in macrophage actin dynamics. Reconstitution of the ancestral version of EsxM in an attenuated modern strain of Mtb altered the migratory mode of infected macrophages, enhancing their motility. In a zebrafish model, full-length EsxM promoted bone disease. The presence of a derived nonsense variant in EsxM throughout the major Mtb lineages 2, 3, and 4 is consistent with a role for EsxM in regulating the extent of dissemination.


Assuntos
Doenças Ósseas , Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculose , Animais , Humanos , Peixe-Zebra , Tuberculose/microbiologia , Macrófagos/microbiologia , Proteínas de Bactérias/genética
3.
Cell ; 184(7): 1757-1774.e14, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33761328

RESUMO

The central pathogen-immune interface in tuberculosis is the granuloma, a complex host immune structure that dictates infection trajectory and physiology. Granuloma macrophages undergo a dramatic transition in which entire epithelial modules are induced and define granuloma architecture. In tuberculosis, relatively little is known about the host signals that trigger this transition. Using the zebrafish-Mycobacterium marinum model, we identify the basis of granuloma macrophage transformation. Single-cell RNA-sequencing analysis of zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected macaques reveal that, even in the presence of robust type 1 immune responses, countervailing type 2 signals associate with macrophage epithelialization. We find that type 2 immune signaling, mediated via stat6, is absolutely required for epithelialization and granuloma formation. In mixed chimeras, stat6 acts cell autonomously within macrophages, where it is required for epithelioid transformation and incorporation into necrotic granulomas. These findings establish the signaling pathway that produces the hallmark structure of mycobacterial infection.


Assuntos
Granuloma/patologia , Imunidade/fisiologia , Infecções por Mycobacterium não Tuberculosas/patologia , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular , Modelos Animais de Doenças , Células Epitelioides/citologia , Células Epitelioides/imunologia , Células Epitelioides/metabolismo , Granuloma/imunologia , Granuloma/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Interferon gama/metabolismo , Interleucina-12/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium marinum/isolamento & purificação , Mycobacterium marinum/fisiologia , Necrose , RNA Guia de Cinetoplastídeos/metabolismo , Receptores de Interleucina-4/antagonistas & inibidores , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/metabolismo , Fator de Transcrição STAT6/antagonistas & inibidores , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
4.
Immunity ; 55(5): 819-821, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35545030

RESUMO

In this issue of Immunity,Gideon et al. (2022) couple sophisticated single-cell analyses with detailed in vivo measurements of Mycobacterium tuberculosis granulomas to define the cellular and transcriptional properties of a successful host immune response during tuberculosis.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Granuloma , Humanos
5.
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
6.
Cell ; 148(3): 434-46, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22304914

RESUMO

Susceptibility to tuberculosis is historically ascribed to an inadequate immune response that fails to control infecting mycobacteria. In zebrafish, we find that susceptibility to Mycobacterium marinum can result from either inadequate or excessive acute inflammation. Modulation of the leukotriene A(4) hydrolase (LTA4H) locus, which controls the balance of pro- and anti-inflammatory eicosanoids, reveals two distinct molecular routes to mycobacterial susceptibility converging on dysregulated TNF levels: inadequate inflammation caused by excess lipoxins and hyperinflammation driven by excess leukotriene B(4). We identify therapies that specifically target each of these extremes. In humans, we identify a single nucleotide polymorphism in the LTA4H promoter that regulates its transcriptional activity. In tuberculous meningitis, the polymorphism is associated with inflammatory cell recruitment, patient survival and response to adjunctive anti-inflammatory therapy. Together, our findings suggest that host-directed therapies tailored to patient LTA4H genotypes may counter detrimental effects of either extreme of inflammation.


Assuntos
Infecções por Mycobacterium/tratamento farmacológico , Infecções por Mycobacterium/imunologia , Tuberculose Meníngea/tratamento farmacológico , Tuberculose Meníngea/imunologia , Animais , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Leucotrieno A4/genética , Leucotrieno A4/imunologia , Leucotrieno B4/genética , Leucotrieno B4/imunologia , Lipoxinas/imunologia , Mitocôndrias/metabolismo , Infecções por Mycobacterium/genética , Mycobacterium marinum , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Transdução de Sinais , Transcrição Gênica , Tuberculose Meníngea/genética , Fator de Necrose Tumoral alfa/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/imunologia
8.
Immunity ; 45(4): 861-876, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760340

RESUMO

Mycobacterium tuberculosis infection in humans triggers formation of granulomas, which are tightly organized immune cell aggregates that are the central structure of tuberculosis. Infected and uninfected macrophages interdigitate, assuming an altered, flattened appearance. Although pathologists have described these changes for over a century, the molecular and cellular programs underlying this transition are unclear. Here, using the zebrafish-Mycobacterium marinum model, we found that mycobacterial granuloma formation is accompanied by macrophage induction of canonical epithelial molecules and structures. We identified fundamental macrophage reprogramming events that parallel E-cadherin-dependent mesenchymal-epithelial transitions. Macrophage-specific disruption of E-cadherin function resulted in disordered granuloma formation, enhanced immune cell access, decreased bacterial burden, and increased host survival, suggesting that the granuloma can also serve a bacteria-protective role. Granuloma macrophages in humans with tuberculosis were similarly transformed. Thus, during mycobacterial infection, granuloma macrophages are broadly reprogrammed by epithelial modules, and this reprogramming alters the trajectory of infection and the associated immune response.


Assuntos
Epitélio/imunologia , Macrófagos/imunologia , Mycobacterium marinum/imunologia , Animais , Caderinas/imunologia , Epitélio/microbiologia , Granuloma/imunologia , Granuloma/microbiologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Peixe-Zebra
9.
Cell ; 140(5): 717-30, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20211140

RESUMO

Exposure to Mycobacterium tuberculosis produces varied early outcomes, ranging from resistance to infection to progressive disease. Here we report results from a forward genetic screen in zebrafish larvae that identify multiple mutant classes with distinct patterns of innate susceptibility to Mycobacterium marinum. A hypersusceptible mutant maps to the lta4h locus encoding leukotriene A(4) hydrolase, which catalyzes the final step in the synthesis of leukotriene B(4) (LTB(4)), a potent chemoattractant and proinflammatory eicosanoid. lta4h mutations confer hypersusceptibility independent of LTB(4) reduction, by redirecting eicosanoid substrates to anti-inflammatory lipoxins. The resultant anti-inflammatory state permits increased mycobacterial proliferation by limiting production of tumor necrosis factor. In humans, we find that protection from both tuberculosis and multibacillary leprosy is associated with heterozygosity for LTA4H polymorphisms that have previously been correlated with differential LTB(4) production. Our results suggest conserved roles for balanced eicosanoid production in vertebrate resistance to mycobacterial infection.


Assuntos
Epóxido Hidrolases/genética , Doenças dos Peixes/genética , Hanseníase/genética , Tuberculose/genética , Animais , Modelos Animais de Doenças , Doenças dos Peixes/imunologia , Predisposição Genética para Doença , Humanos , Hanseníase/imunologia , Tuberculose/imunologia , Peixe-Zebra
10.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33658385

RESUMO

Adjunctive treatment with antiinflammatory corticosteroids like dexamethasone increases survival in tuberculosis meningitis. Dexamethasone responsiveness associates with a C/T variant in Leukotriene A4 Hydrolase (LTA4H), which regulates expression of the proinflammatory mediator leukotriene B4 (LTB4). TT homozygotes, with increased expression of LTA4H, have the highest survival when treated with dexamethasone and the lowest survival without. While the T allele is present in only a minority of the world's population, corticosteroids confer modest survival benefit worldwide. Using Bayesian methods, we examined how pretreatment levels of cerebrospinal fluid proinflammatory cytokines affect survival in dexamethasone-treated tuberculous meningitis. LTA4H TT homozygosity was associated with global cytokine increases, including tumor necrosis factor. Association between higher cytokine levels and survival extended to non-TT patients, suggesting that other genetic variants may also induce dexamethasone-responsive pathological inflammation. These findings warrant studies that tailor dexamethasone therapy to pretreatment cerebrospinal fluid cytokine concentrations, while searching for additional genetic loci shaping the inflammatory milieu.


Assuntos
Citocinas/líquido cefalorraquidiano , Dexametasona/administração & dosagem , Epóxido Hidrolases/genética , Variação Genética , Tuberculose Meníngea , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Taxa de Sobrevida , Tuberculose Meníngea/líquido cefalorraquidiano , Tuberculose Meníngea/tratamento farmacológico , Tuberculose Meníngea/genética , Tuberculose Meníngea/mortalidade
11.
J Cell Sci ; 134(8)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33722979

RESUMO

CRISPR/Cas9-based tissue-specific knockout techniques are essential for probing the functions of genes in embryonic development and disease using zebrafish. However, the lack of capacity to perform gene-specific rescue or live imaging in the tissue-specific knockout background has limited the utility of this approach. Here, we report a robust and flexible gateway system for tissue-specific gene inactivation in neutrophils. Using a transgenic fish line with neutrophil-restricted expression of Cas9 and ubiquitous expression of single guide (sg)RNAs targeting rac2, specific disruption of the rac2 gene in neutrophils is achieved. Transient expression of sgRNAs targeting rac2 or cdk2 in the neutrophil-restricted Cas9 line also results in significantly decreased cell motility. Re-expressing sgRNA-resistant rac2 or cdk2 genes restores neutrophil motility in the corresponding knockout background. Moreover, active Rac and force-bearing F-actins localize to both the cell front and the contracting tail during neutrophil interstitial migration in an oscillating fashion that is disrupted when rac2 is knocked out. Together, our work provides a potent tool that can be used to advance the utility of zebrafish in identifying and characterizing gene functions in a tissue-specific manner.


Assuntos
Neutrófilos , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Sistemas CRISPR-Cas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Neutrófilos/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo
12.
Proc Natl Acad Sci U S A ; 116(34): 16961-16970, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31391308

RESUMO

Intestinal epithelial cell (IEC) shedding is a fundamental response to intestinal damage, yet underlying mechanisms and functions have been difficult to define. Here we model chronic intestinal damage in zebrafish larvae using the nonsteroidal antiinflammatory drug (NSAID) Glafenine. Glafenine induced the unfolded protein response (UPR) and inflammatory pathways in IECs, leading to delamination. Glafenine-induced inflammation was augmented by microbial colonization and associated with changes in intestinal and environmental microbiotas. IEC shedding was a UPR-dependent protective response to Glafenine that restricts inflammation and promotes animal survival. Other NSAIDs did not induce IEC delamination; however, Glafenine also displays off-target inhibition of multidrug resistance (MDR) efflux pumps. We found a subset of MDR inhibitors also induced IEC delamination, implicating MDR efflux pumps as cellular targets underlying Glafenine-induced enteropathy. These results implicate IEC delamination as a protective UPR-mediated response to chemical injury, and uncover an essential role for MDR efflux pumps in intestinal homeostasis.


Assuntos
Anti-Inflamatórios não Esteroides , Enterócitos/metabolismo , Microbioma Gastrointestinal , Glafenina/efeitos adversos , Enteropatias , Peixe-Zebra , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacologia , Enterócitos/microbiologia , Enterócitos/patologia , Glafenina/farmacologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Enteropatias/induzido quimicamente , Enteropatias/metabolismo , Enteropatias/microbiologia , Enteropatias/patologia , Peixe-Zebra/metabolismo , Peixe-Zebra/microbiologia
13.
Clin Infect Dis ; 72(12): e931-e937, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33136139

RESUMO

BACKGROUND: Nontuberculous mycobacteria (NTM) are a rare cause of infectious tenosynovitis of the upper extremity. Using molecular methods, clinical microbiology laboratories are increasingly reporting identification down to the species level. Improved methods for speciation are revealing new insights into the clinical and epidemiologic features of rare NTM infections. METHODS: We encountered 3 cases of epidemiologically linked upper extremity NTM tenosynovitis associated with exposure to hurricane-damaged wood. We conducted whole-genome sequencing to assess isolate relatedness followed by a literature review of NTM infections that involved the upper extremity. RESULTS: Despite shared epidemiologic risk, the cases were caused by 3 distinct organisms. Two cases were rare infections caused by closely related but distinct species within the Mycobacterium terrae complex that could not be differentiated by traditional methods. The third case was caused by Mycobacterium intracellulare. An updated literature review that focused on research that used modern molecular speciation methods found that several species within the M. terrae complex are increasingly reported as a cause of upper extremity tenosynovitis, often in association with environmental exposures. CONCLUSIONS: These cases illustrate the importance of molecular methods for speciating phenotypically similar NTM, as well as the limitations of laboratory-based surveillance in detecting point-source outbreaks when the source is environmental and may involve multiple organisms.


Assuntos
Tempestades Ciclônicas , Infecções por Mycobacterium não Tuberculosas , Tenossinovite , Humanos , Infecções por Mycobacterium não Tuberculosas/epidemiologia , Complexo Mycobacterium avium , Micobactérias não Tuberculosas/genética , Tenossinovite/epidemiologia
14.
Nat Methods ; 15(12): 1098-1107, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30504889

RESUMO

A central and critical structure in tuberculosis, the mycobacterial granuloma consists of highly organized immune cells, including macrophages that drive granuloma formation through a characteristic epithelioid transformation. Difficulties in imaging within intact animals and caveats associated with in vitro assembly models have severely limited the study and experimental manipulation of mature granulomas. Here we describe a new ex vivo culture technique, wherein mature, fully organized zebrafish granulomas are microdissected and maintained in three-dimensional (3D) culture. This approach enables high-resolution microscopy of granuloma macrophage dynamics, including epithelioid macrophage motility and granuloma consolidation, while retaining key bacterial and host characteristics. Using mass spectrometry, we find active production of key phosphotidylinositol species identified previously in human granulomas. We also describe a method to transfect isolated granulomas, enabling genetic manipulation, and provide proof-of-concept for host-directed small-molecule screens, identifying protein kinase C (PKC) signaling as an important regulator of granuloma macrophage organization.


Assuntos
Granuloma/patologia , Processamento de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Macrófagos/patologia , Tuberculose/patologia , Animais , Animais Geneticamente Modificados , Granuloma/tratamento farmacológico , Granuloma/microbiologia , Indóis/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Maleimidas/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , Peixe-Zebra
15.
PLoS Pathog ; 15(3): e1007381, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30845179

RESUMO

The intestinal microbiota influences the development and function of myeloid lineages such as neutrophils, but the underlying molecular mechanisms are unresolved. Using gnotobiotic zebrafish, we identified the immune effector Serum amyloid A (Saa) as one of the most highly induced transcripts in digestive tissues following microbiota colonization. Saa is a conserved secreted protein produced in the intestine and liver with described effects on neutrophils in vitro, however its in vivo functions remain poorly defined. We engineered saa mutant zebrafish to test requirements for Saa on innate immunity in vivo. Zebrafish mutant for saa displayed impaired neutrophil responses to wounding but augmented clearance of pathogenic bacteria. At baseline, saa mutants exhibited moderate neutrophilia and altered neutrophil tissue distribution. Molecular and functional analyses of isolated neutrophils revealed that Saa suppresses expression of pro-inflammatory markers and bactericidal activity. Saa's effects on neutrophils depended on microbiota colonization, suggesting this protein mediates the microbiota's effects on host innate immunity. To test tissue-specific roles of Saa on neutrophil function, we over-expressed saa in the intestine or liver and found that sufficient to partially complement neutrophil phenotypes observed in saa mutants. These results indicate Saa produced by the intestine in response to microbiota serves as a systemic signal to neutrophils to restrict aberrant activation, decreasing inflammatory tone and bacterial killing potential while simultaneously enhancing their ability to migrate to wounds.


Assuntos
Ativação de Neutrófilo/fisiologia , Proteína Amiloide A Sérica/fisiologia , Peixe-Zebra/microbiologia , Animais , Imunidade Inata/fisiologia , Intestinos , Fígado , Microbiota , Neutrófilos/fisiologia , Proteína Amiloide A Sérica/metabolismo , Transdução de Sinais
16.
Nat Chem Biol ; 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898167
17.
Nature ; 517(7536): 612-5, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25470057

RESUMO

Pathogenic mycobacteria induce the formation of complex cellular aggregates called granulomas that are the hallmark of tuberculosis. Here we examine the development and consequences of vascularization of the tuberculous granuloma in the zebrafish-Mycobacterium marinum infection model, which is characterized by organized granulomas with necrotic cores that bear striking resemblance to those of human tuberculosis. Using intravital microscopy in the transparent larval zebrafish, we show that granuloma formation is intimately associated with angiogenesis. The initiation of angiogenesis in turn coincides with the generation of local hypoxia and transcriptional induction of the canonical pro-angiogenic molecule Vegfaa. Pharmacological inhibition of the Vegf pathway suppresses granuloma-associated angiogenesis, reduces infection burden and limits dissemination. Moreover, anti-angiogenic therapies synergize with the first-line anti-tubercular antibiotic rifampicin, as well as with the antibiotic metronidazole, which targets hypoxic bacterial populations. Our data indicate that mycobacteria induce granuloma-associated angiogenesis, which promotes mycobacterial growth and increases spread of infection to new tissue sites. We propose the use of anti-angiogenic agents, now being used in cancer regimens, as a host-targeting tuberculosis therapy, particularly in extensively drug-resistant disease for which current antibiotic regimens are largely ineffective.


Assuntos
Inibidores da Angiogênese/farmacologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/efeitos dos fármacos , Mycobacterium marinum/crescimento & desenvolvimento , Neovascularização Patológica/microbiologia , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/microbiologia , Inibidores da Angiogênese/uso terapêutico , Animais , Antibióticos Antituberculose/farmacologia , Carga Bacteriana/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Granuloma/tratamento farmacológico , Granuloma/metabolismo , Granuloma/microbiologia , Granuloma/patologia , Hipóxia/metabolismo , Hipóxia/microbiologia , Hipóxia/patologia , Larva/efeitos dos fármacos , Larva/microbiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/metabolismo , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium marinum/patogenicidade , Neovascularização Patológica/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , Tuberculose/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/crescimento & desenvolvimento
18.
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
19.
Proc Natl Acad Sci U S A ; 114(37): E7746-E7755, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28827342

RESUMO

Risk, severity, and outcome of infection depend on the interplay of pathogen virulence and host susceptibility. Systematic identification of genetic susceptibility to infection is being undertaken through genome-wide association studies, but how to expeditiously move from genetic differences to functional mechanisms is unclear. Here, we use genetic association of molecular, cellular, and human disease traits and experimental validation to demonstrate that genetic variation affects expression of VAC14, a phosphoinositide-regulating protein, to influence susceptibility to Salmonella enterica serovar Typhi (S Typhi) infection. Decreased VAC14 expression increased plasma membrane cholesterol, facilitating Salmonella docking and invasion. This increased susceptibility at the cellular level manifests as increased susceptibility to typhoid fever in a Vietnamese population. Furthermore, treating zebrafish with a cholesterol-lowering agent, ezetimibe, reduced susceptibility to S Typhi. Thus, coupling multiple genetic association studies with mechanistic dissection revealed how VAC14 regulates Salmonella invasion and typhoid fever susceptibility and may open doors to new prophylactic/therapeutic approaches.


Assuntos
Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Salmonella typhi/genética , Linhagem Celular Tumoral , Colesterol/genética , Colesterol/metabolismo , Ezetimiba , Variação Genética/genética , Estudo de Associação Genômica Ampla , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Polimorfismo de Nucleotídeo Único , Salmonella/genética , Salmonella/patogenicidade , Salmonella typhi/metabolismo , Salmonella typhi/patogenicidade , Febre Tifoide/metabolismo , Febre Tifoide/fisiopatologia , Virulência/genética
20.
J Infect Dis ; 220(3): 524-534, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30877311

RESUMO

BACKGROUND: Infection-induced thrombocytosis is a clinically important complication of tuberculosis infection. Recent studies have highlighted the utility of aspirin as a host-directed therapy modulating the inflammatory response to infection but have not investigated the possibility that the effect of aspirin is related to an antiplatelet mode of action. METHODS: In this study, we utilize the zebrafish-Mycobacterium marinum model to show mycobacteria drive host hemostasis through the formation of granulomas. Treatment of infected zebrafish with aspirin markedly reduced mycobacterial burden. This effect is reproduced by treatment with platelet-specific glycoprotein IIb/IIIa inhibitors demonstrating a detrimental role for infection-induced thrombocyte activation. RESULTS: We find that the reduction in mycobacterial burden is dependent on macrophages and granuloma formation, providing the first in vivo experimental evidence that infection-induced platelet activation compromises protective host immunity to mycobacterial infection. CONCLUSIONS: Our study illuminates platelet activation as an efficacious target of aspirin, a widely available and affordable host-directed therapy candidate for tuberculosis.


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/imunologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium marinum/imunologia , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia , Animais , Proteínas de Bactérias/imunologia , Modelos Animais de Doenças , Granuloma/tratamento farmacológico , Granuloma/imunologia , Granuloma/microbiologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Tuberculose/tratamento farmacológico , Tuberculose/imunologia , Tuberculose/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA