Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mBio ; 15(4): e0335723, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38445877

RESUMEN

Bacterial pathogens use protein secretion systems to transport virulence factors and regulate gene expression. Among pathogenic mycobacteria, including Mycobacterium tuberculosis and Mycobacterium marinum, the ESAT-6 system 1 (ESX-1) secretion is crucial for host interaction. Secretion of protein substrates by the ESX-1 secretion system disrupts phagosomes, allowing mycobacteria cytoplasmic access during macrophage infections. Deletion or mutation of the ESX-1 system attenuates mycobacterial pathogens. Pathogenic mycobacteria respond to the presence or absence of the ESX-1 system in the cytoplasmic membrane by altering transcription. Under laboratory conditions, the EspM repressor and WhiB6 activator control transcription of specific ESX-1-responsive genes, including the ESX-1 substrate genes. However, deleting the espM or whiB6 gene does not phenocopy the deletion of the ESX-1 substrate genes during macrophage infection by M. marinum. In this study, we identified EspN, a critical transcription factor whose activity is masked by the EspM repressor under laboratory conditions. In the absence of EspM, EspN activates transcription of whiB6 and ESX-1 genes during both laboratory growth and macrophage infection. EspN is also independently required for M. marinum growth within and cytolysis of macrophages, similar to the ESX-1 genes, and for disease burden in a zebrafish larval model of infection. These findings suggest that EspN and EspM coordinate to counterbalance the regulation of the ESX-1 system and support mycobacterial pathogenesis.IMPORTANCEPathogenic mycobacteria, which are responsible for tuberculosis and other long-term diseases, use the ESX-1 system to transport proteins that control the host response to infection and promote bacterial survival. In this study, we identify an undescribed transcription factor that controls the expression of ESX-1 genes and is required for both macrophage and animal infection. However, this transcription factor is not the primary regulator of ESX-1 genes under standard laboratory conditions. These findings identify a critical transcription factor that likely controls expression of a major virulence pathway during infection, but whose effect is not detectable with standard laboratory strains and growth conditions.


Asunto(s)
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Sistemas de Secreción Tipo VII , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Tipo VII/genética , Sistemas de Secreción Tipo VII/metabolismo , Pez Cebra , Tuberculosis/microbiología , Mycobacterium tuberculosis/metabolismo , Mycobacterium marinum/metabolismo
2.
bioRxiv ; 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36824794

RESUMEN

Bacterial pathogens use protein secretion systems to translocate virulence factors into the host and to control bacterial gene expression. The ESX-1 (ESAT-6 system 1) secretion system facilitates disruption of the macrophage phagosome during infection, enabling access to the cytoplasm, and regulates widespread gene expression in the mycobacterial cell. The transcription factors contributing to the ESX-1 transcriptional network during mycobacterial infection are not known. We showed that the EspM and WhiB6 transcription factors regulate the ESX-1 transcriptional network in vitro but are dispensable for macrophage infection by Mycobacterium marinum . In this study, we used our understanding of the ESX-1 system to identify EspN, a critical transcription factor that controls expression of the ESX-1 genes during infection, but whose effect is not detectable under standard laboratory growth conditions. Under laboratory conditions, EspN activity is masked by the EspM repressor. In the absence of EspM, we found that EspN is required for ESX-1 function because it activates expression of the whiB6 transcription factor gene, and specific ESX-1 substrate and secretory component genes. Unlike the other transcription factors that regulate ESX-1, EspN is required for M. marinum growth within and cytolysis of macrophages, and for disease burden in a zebrafish larval model of infection. These findings demonstrate that EspN is an infection-dependent regulator of the ESX-1 transcriptional network, which is essential for mycobacterial pathogenesis. Moreover, our findings suggest that ESX-1 expression is controlled by a genetic switch that responds to host specific signals. Importance: Pathogenic mycobacteria cause acute and long-term diseases, including human tuberculosis. The ESX-1 system transports proteins that control the host response to infection and promotes bacterial survival. Although ESX-1 transports proteins, it also controls gene expression in the bacteria. In this study, we identify an undescribed transcription factor that controls the expression of ESX-1 genes, and is required for both macrophage and animal infection. However, this transcription factor is not the primary regulator of ESX-1 genes under standard laboratory conditions. These findings identify a critical transcription factor that controls expression of a major virulence pathway during infection, but whose effect is not detectable with standard laboratory strains and growth conditions.

3.
Cell Rep ; 41(11): 111817, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36516756

RESUMEN

During mycobacterial infections, pathogenic mycobacteria manipulate both host immune and stromal cells to establish and maintain a productive infection. In humans, non-human primates, and zebrafish models of infection, pathogenic mycobacteria produce and modify the specialized lipid trehalose 6,6'-dimycolate (TDM) in the bacterial cell envelope to drive host angiogenesis toward the site of forming granulomas, leading to enhanced bacterial growth. Here, we use the zebrafish-Mycobacterium marinum infection model to define the signaling basis of the host angiogenic response. Through intravital imaging and cell-restricted peptide-mediated inhibition, we identify macrophage-specific activation of NFAT signaling as essential to TDM-mediated angiogenesis in vivo. Exposure of cultured human cells to Mycobacterium tuberculosis results in robust induction of VEGFA, which is dependent on a signaling pathway downstream of host TDM detection and culminates in NFATC2 activation. As granuloma-associated angiogenesis is known to serve bacterial-beneficial roles, these findings identify potential host targets to improve tuberculosis disease outcomes.


Asunto(s)
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animales , Humanos , Pez Cebra/microbiología , Macrófagos/metabolismo , Transducción de Señal , Granuloma/patología , Factores de Transcripción NFATC/metabolismo
4.
Cell ; 185(24): 4507-4525.e18, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36356582

RESUMEN

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.


Asunto(s)
Enfermedades Óseas , Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animales , Humanos , Pez Cebra , Tuberculosis/microbiología , Macrófagos/microbiología , Proteínas Bacterianas/genética
5.
Cell ; 185(20): 3720-3738.e13, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36103894

RESUMEN

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.


Asunto(s)
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animales , Mycobacterium tuberculosis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Pez Cebra
6.
Dis Model Mech ; 15(8)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36037003

RESUMEN

During the current COVID-19 pandemic, there has been renewed scientific and public focus on understanding the pathogenesis of infectious diseases and investigating vaccines and therapies to combat them. In addition to the tragic toll of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we also recognize increased threats from antibiotic-resistant bacterial strains, the effects of climate change on the prevalence and spread of human pathogens, and the recalcitrance of other infectious diseases - including tuberculosis, malaria, human immunodeficiency virus (HIV) and fungal infections - that continue to cause millions of deaths annually. Large amounts of funding have rightly been redirected toward vaccine development and clinical trials for COVID-19, but we must continue to pursue fundamental and translational research on other pathogens and host immunity. Now more than ever, we need to support the next generation of researchers to develop and utilize models of infectious disease that serve as engines of discovery, innovation and therapy.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Tuberculosis , Enfermedades Transmisibles/epidemiología , Humanos , Pandemias , SARS-CoV-2
7.
Immunity ; 55(5): 819-821, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545030

RESUMEN

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.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Granuloma , Humanos
8.
Pathog Dis ; 80(1)2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35438161

RESUMEN

Mycobacterial granuloma formation involves significant stromal remodeling including the growth of leaky, granuloma-associated vasculature. These permeable blood vessels aid mycobacterial growth, as antiangiogenic or vascular normalizing therapies are beneficial host-directed therapies in preclinical models of tuberculosis across host-mycobacterial pairings. Using the zebrafish-Mycobacterium marinum infection model, we demonstrate that vascular normalization by inhibition of vascular endothelial protein tyrosine phosphatase (VE-PTP) decreases granuloma hypoxia, the opposite effect of hypoxia-inducing antiangiogenic therapy. Inhibition of VE-PTP decreased neutrophil recruitment to granulomas in adult and larval zebrafish, and decreased the proportion of neutrophils that extravasated distal to granulomas. Furthermore, VE-PTP inhibition increased the accumulation of T cells at M. marinum granulomas. Our study provides evidence that, similar to the effect in solid tumors, vascular normalization during mycobacterial infection increases the T cell:neutrophil ratio in lesions which may be correlates of protective immunity.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas , Mycobacterium marinum , Mycobacterium , Animales , Permeabilidad Capilar , Modelos Animales de Enfermedad , Granuloma , Hipoxia , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/metabolismo , Neutrófilos , Pez Cebra/microbiología
9.
Cell Syst ; 12(11): 1021-1022, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34793699

RESUMEN

Improved therapies for tuberculosis will require the careful revision of complex, multi-drug regimens. In this issue of Cell Systems, Larkins-Ford et al. integrate extensive dose-response measurements of drug combinations, in vivo animal data, and computational analysis to provide a new predictive framework for the prioritization of specific antitubercular drug regimens.


Asunto(s)
Antituberculosos , Tuberculosis , Animales , Antituberculosos/uso terapéutico , Combinación de Medicamentos , Tuberculosis/tratamiento farmacológico
10.
Cell ; 184(7): 1757-1774.e14, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33761328

RESUMEN

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.


Asunto(s)
Granuloma/patología , Inmunidad/fisiología , Infecciones por Mycobacterium no Tuberculosas/patología , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Células Epitelioides/citología , Células Epitelioides/inmunología , Células Epitelioides/metabolismo , Granuloma/inmunología , Granuloma/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/aislamiento & purificación , Mycobacterium marinum/fisiología , Necrosis , ARN Guía de Kinetoplastida/metabolismo , Receptores de Interleucina-4/antagonistas & inhibidores , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/metabolismo , Factor de Transcripción STAT6/antagonistas & inhibidores , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
11.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33658385

RESUMEN

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.


Asunto(s)
Citocinas/líquido cefalorraquídeo , Dexametasona/administración & dosificación , Epóxido Hidrolasas/genética , Variación Genética , Tuberculosis Meníngea , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Tasa de Supervivencia , Tuberculosis Meníngea/líquido cefalorraquídeo , Tuberculosis Meníngea/tratamiento farmacológico , Tuberculosis Meníngea/genética , Tuberculosis Meníngea/mortalidad
12.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33645551

RESUMEN

Nearly 140 years after Robert Koch discovered Mycobacterium tuberculosis, tuberculosis (TB) remains a global threat and a deadly human pathogen. M. tuberculosis is notable for complex host-pathogen interactions that lead to poorly understood disease states ranging from latent infection to active disease. Additionally, multiple pathologies with a distinct local milieu (bacterial burden, antibiotic exposure, and host response) can coexist simultaneously within the same subject and change independently over time. Current tools cannot optimally measure these distinct pathologies or the spatiotemporal changes. Next-generation molecular imaging affords unparalleled opportunities to visualize infection by providing holistic, 3D spatial characterization and noninvasive, temporal monitoring within the same subject. This rapidly evolving technology could powerfully augment TB research by advancing fundamental knowledge and accelerating the development of novel diagnostics, biomarkers, and therapeutics.


Asunto(s)
Imagen Molecular , Mycobacterium tuberculosis/metabolismo , Tuberculosis/diagnóstico por imagen , Tuberculosis/metabolismo , Animales , Biomarcadores/metabolismo , Humanos
13.
J Cell Sci ; 134(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33722979

RESUMEN

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.


Asunto(s)
Neutrófilos , Pez Cebra , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Neutrófilos/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Unión al GTP rac/metabolismo
14.
Clin Infect Dis ; 72(12): e931-e937, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33136139

RESUMEN

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.


Asunto(s)
Tormentas Ciclónicas , Infecciones por Mycobacterium no Tuberculosas , Tenosinovitis , Humanos , Infecciones por Mycobacterium no Tuberculosas/epidemiología , Complejo Mycobacterium avium , Micobacterias no Tuberculosas/genética , Tenosinovitis/epidemiología
15.
PLoS One ; 15(5): e0232251, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32407412

RESUMEN

Lipids represent an important source of nutrition for infecting mycobacteria, accumulating within the necrotic core of granulomas and present in foamy macrophages associated with mycobacterial infection. In order to better understand the timing, process and importance of lipid accumulation, we developed methods for direct in vivo visualization and quantification of this process using the zebrafish-M. marinum larval model of infection. We find that neutral lipids accumulate cell-autonomously in mycobacterium-infected macrophages in vivo during early infection, with detectable levels of accumulation by two days post-infection. Treatment with ezetimibe, an FDA-approved drug, resulted in decreased levels of free cholesterol and neutral lipids, and a reduction of bacterial growth in vivo. The effect of ezetimibe in reducing bacterial growth was dependent on the mce4 operon, a key bacterial determinant of lipid utilization. Thus, in vivo, lipid accumulation can occur cell-autonomously at early timepoints of mycobacterial infection, and limitation of this process results in decreased bacterial burden.


Asunto(s)
Metabolismo de los Lípidos , Mycobacterium marinum/crecimiento & desarrollo , Ezetimiba/farmacología , Macrófagos/metabolismo , Macrófagos/microbiología , Mutación , Mycobacterium marinum/efectos de los fármacos , Mycobacterium marinum/genética , Mycobacterium marinum/fisiología , Operón/genética
16.
J Leukoc Biol ; 107(2): 171-173, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31667877

RESUMEN

Discussion on an unusual role for a cxcr3 receptor, in which it antagonizes a paralogous receptor to limit macrophage migration.


Asunto(s)
Infecciones , Receptores CXCR3 , Animales , Movimiento Celular , Macrófagos , Pez Cebra
17.
Proc Natl Acad Sci U S A ; 116(34): 16961-16970, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31391308

RESUMEN

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.


Asunto(s)
Antiinflamatorios no Esteroideos , Enterocitos/metabolismo , Microbioma Gastrointestinal , Glafenina/efectos adversos , Enfermedades Intestinales , Pez Cebra , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Enterocitos/microbiología , Enterocitos/patología , Glafenina/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Enfermedades Intestinales/inducido químicamente , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/patología , Pez Cebra/metabolismo , Pez Cebra/microbiología
18.
Front Immunol ; 10: 528, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30967867

RESUMEN

Over the past 200 years, tuberculosis (TB) has caused more deaths than any other infectious disease, likely infecting more people than it has at any other time in human history. Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, is an obligate human pathogen that has evolved through the millennia to become an archetypal human-adapted pathogen. This review focuses on the evolutionary framework by which Mtb emerged as a specialized human pathogen and applies this perspective to the emergence of specific lineages that drive global TB burden. We consider how evolutionary pressures, including transmission dynamics, host tolerance, and human population patterns, may have shaped the evolution of diverse mycobacterial genomes.


Asunto(s)
Evolución Biológica , Interacciones Huésped-Patógeno/inmunología , Tolerancia Inmunológica , Mycobacterium tuberculosis/fisiología , Tuberculosis/inmunología , Humanos
20.
J Infect Dis ; 220(3): 524-534, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-30877311

RESUMEN

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.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/inmunología , Pez Cebra/inmunología , Pez Cebra/microbiología , Animales , Proteínas Bacterianas/inmunología , Modelos Animales de Enfermedad , Granuloma/tratamiento farmacológico , Granuloma/inmunología , Granuloma/microbiología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Tuberculosis/tratamiento farmacológico , Tuberculosis/inmunología , Tuberculosis/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...