Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Trials ; 24(1): 435, 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37370174

RESUMEN

BACKGROUND: The duration and regimen of tuberculosis (TB) treatment is currently based predominantly on whether the M. tuberculosis (Mtb) strain is drug-sensitive (DS) or multidrug-resistant (MDR) with doses adjusted by patients' weight only. The systematic stratification of patients for personalized treatment does not exist for TB. As each TB case is different, individualized treatment regimens should be applied to obtain better outcomes. In this scenario, novel therapeutic approaches are urgently needed to (1) improve outcomes and (2) shorten treatment duration, and host-directed therapies (HDT) might be the best solution. Within HDT, repurposed drugs represent a shortcut in drug development and can be implemented at the short term. As hyperinflammation is associated with worse outcomes, HDT with an anti-inflammatory effect might improve outcomes by reducing tissue damage and thus the risk of permanent sequelae. METHODS: SMA-TB is a multicentre randomized, phase IIB, placebo-controlled, three-arm, double-blinded clinical trial (CT) that has been designed in the context of the EC-funded SMA-TB Project ( www.smatb.eu ) in which we propose to use 2 common non-steroidal anti-inflammatory drugs (NSAID), acetylsalicylic acid (ASA) and ibuprofen (Ibu), as an HDT for use as adjunct therapy added to, and compared with, the standard of care (SoC) World Health Organization (WHO)-recommended TB regimen in TB patients. A total of 354 South African and Georgian adults diagnosed with confirmed pulmonary TB will be randomized into SoC TB treatment + placebo, SoC + acetylsalicylic acid or SoC + ibuprofen. DISCUSSION: SMA-TB will provide proof of concept of the HDT as a co-adjuvant treatment and identify the suitability of the intervention for different population groups (different epidemiological settings and drug susceptibility) in the reduction of tissue damage and risk of bad outcomes for TB patients. This regimen potentially will be more effective and targeted: organ saving, reducing tissue damage and thereby decreasing the length of treatment and sequelae, increasing cure rates and pathogen clearance and decreasing transmission rates. It will result in better clinical practice, care management and increased well-being of TB patients. TRIAL REGISTRATION: Clinicaltrials.gov NCT04575519. Registered on October 5, 2020.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis Pulmonar , Adulto , Humanos , Antiinflamatorios/uso terapéutico , Antituberculosos/efectos adversos , Aspirina/efectos adversos , Ibuprofeno/efectos adversos , Estudios Multicéntricos como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento , Tuberculosis Pulmonar/diagnóstico , Organización Mundial de la Salud , Ensayos Clínicos Fase II como Asunto
2.
Microorganisms ; 9(1)2021 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-33406734

RESUMEN

Glycans display increasingly recognized roles in pathological contexts, however, their impact in the host-pathogen interplay in many infectious diseases remains largely unknown. This is the case for tuberculosis (TB), one of the ten most fatal diseases worldwide, caused by infection of the bacteria Mycobacterium tuberculosis. We have recently reported that perturbing the core-2 O-glycans biosynthetic pathway increases the host susceptibility to M. tuberculosis infection, by disrupting the neutrophil homeostasis and enhancing lung pathology. In the present study, we show an increased expression of the sialylated glycan structure Sialyl-Lewis X (SLeX) in the lung epithelium upon M. tuberculosis infection. This increase in SLeX glycan epitope is accompanied by an altered lung tissue transcriptomic signature, with up-regulation of genes codifying enzymes that are involved in the SLeX core-2 O-glycans biosynthetic pathway. This study provides novel insights into previously unappreciated molecular mechanisms involving glycosylation, which modulate the host response to M. tuberculosis infection, possibly contributing to shape TB disease outcome.

3.
Nat Commun ; 11(1): 5566, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33149141

RESUMEN

Tuberculosis (TB) is a leading cause of mortality due to infectious disease, but the factors determining disease progression are unclear. Transcriptional signatures associated with type I IFN signalling and neutrophilic inflammation were shown to correlate with disease severity in mouse models of TB. Here we show that similar transcriptional signatures correlate with increased bacterial loads and exacerbate pathology during Mycobacterium tuberculosis infection upon GM-CSF blockade. Loss of GM-CSF signalling or genetic susceptibility to TB (C3HeB/FeJ mice) result in type I IFN-induced neutrophil extracellular trap (NET) formation that promotes bacterial growth and promotes disease severity. Consistently, NETs are present in necrotic lung lesions of TB patients responding poorly to antibiotic therapy, supporting the role of NETs in a late stage of TB pathogenesis. Our findings reveal an important cytokine-based innate immune effector network with a central role in determining the outcome of M. tuberculosis infection.


Asunto(s)
Trampas Extracelulares/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interferón Tipo I/metabolismo , Pulmón/microbiología , Mycobacterium tuberculosis/inmunología , Neutrófilos/inmunología , Neumonía/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Bases de Datos Genéticas , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Interferón Tipo I/genética , Interferón gamma/genética , Interferón gamma/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Neumonía/genética , Neumonía/metabolismo , Neumonía/patología , RNA-Seq , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Tuberculosis Pulmonar/sangre , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/microbiología
4.
Nat Commun ; 11(1): 1949, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32327653

RESUMEN

Genetic diversity of Mycobacterium tuberculosis affects immune responses and clinical outcomes of tuberculosis (TB). However, how bacterial diversity orchestrates immune responses to direct distinct TB severities is unknown. Here we study 681 patients with pulmonary TB and show that M. tuberculosis isolates from cases with mild disease consistently induce robust cytokine responses in macrophages across multiple donors. By contrast, bacteria from patients with severe TB do not do so. Secretion of IL-1ß is a good surrogate of the differences observed, and thus to classify strains as probable drivers of different TB severities. Furthermore, we demonstrate that M. tuberculosis isolates that induce low levels of IL-1ß production can evade macrophage cytosolic surveillance systems, including cGAS and the inflammasome. Isolates exhibiting this evasion strategy carry candidate mutations, generating sigA recognition boxes or affecting components of the ESX-1 secretion system. Therefore, we provide evidence that M. tuberculosis strains manipulate host-pathogen interactions to drive variable TB severities.


Asunto(s)
Citosol/inmunología , Interleucina-1beta/metabolismo , Mycobacterium tuberculosis/patogenicidad , Transducción de Señal/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Proteínas Bacterianas/genética , Células Cultivadas , Citocinas/metabolismo , Femenino , Genoma Bacteriano/genética , Humanos , Evasión Inmune , Inmunomodulación , Inflamasomas/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Mutación , Mycobacterium tuberculosis/clasificación , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/aislamiento & purificación , Filogenia , Polimorfismo de Nucleótido Simple , Tuberculosis Pulmonar/microbiología , Virulencia/genética
5.
Mucosal Immunol ; 13(5): 836-848, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32203062

RESUMEN

Modulation of immunity and disease by glycans is increasingly recognized. However, how host glycosylation shapes and is shaped by tuberculosis remains poorly understood. We show that deficiency in the glucosaminyl (N-acetyl) transferase 1 (Gcnt1), a key enzyme for core-2 O-glycans biosynthesis, drives susceptibility to Mycobacterium tuberculosis infection. The increased susceptibility of Gcnt1 deficient mice was characterized by extensive lung immune pathology, mechanistically related to neutrophils. Uninfected Gcnt1 deficient mice presented bone marrow, blood and lung neutrophilia, which further increased with infection. Blood neutrophilia required Gcnt1 deficiency in the hematopoietic compartment, relating with enhanced granulopoiesis, but normal cellular egress from the bone marrow. Interestingly, for the blood neutrophilia to translate into susceptibility to M. tuberculosis infection, Gnct1 deficiency in the stroma was also necessary. Complete Gcnt1 deficiency associated with increased lung expression of the neutrophil chemoattractant CXCL2. Lastly, we demonstrate that the transcript levels of various glycosyltransferase-encoding genes were altered in whole blood of active tuberculosis patients and that sialyl Lewis x, a glycan widely present in human neutrophils, was detected in the lung of tuberculosis patients. Our findings reveal a previously unappreciated link between Gcnt1, neutrophilia and susceptibility to M. tuberculosis infection, uncovering new players balancing the immune response in tuberculosis.


Asunto(s)
Predisposición Genética a la Enfermedad , Mycobacterium tuberculosis , N-Acetilglucosaminiltransferasas/deficiencia , Neutrófilos/inmunología , Neutrófilos/metabolismo , Tuberculosis/etiología , Tuberculosis/metabolismo , Animales , Carga Bacteriana , Biomarcadores , Modelos Animales de Enfermedad , Activación Enzimática , Regulación de la Expresión Génica , Glicosilación , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Neutrófilos/patología , Tasa de Supervivencia , Tuberculosis/diagnóstico , Tuberculosis/mortalidad
6.
Nat Immunol ; 21(4): 464-476, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205882

RESUMEN

Although mouse infection models have been extensively used to study the host response to Mycobacterium tuberculosis, their validity in revealing determinants of human tuberculosis (TB) resistance and disease progression has been heavily debated. Here, we show that the modular transcriptional signature in the blood of susceptible mice infected with a clinical isolate of M. tuberculosis resembles that of active human TB disease, with dominance of a type I interferon response and neutrophil activation and recruitment, together with a loss in B lymphocyte, natural killer and T cell effector responses. In addition, resistant but not susceptible strains of mice show increased lung B cell, natural killer and T cell effector responses in the lung upon infection. Notably, the blood signature of active disease shared by mice and humans is also evident in latent TB progressors before diagnosis, suggesting that these responses both predict and contribute to the pathogenesis of progressive M. tuberculosis infection.


Asunto(s)
Transcriptoma/inmunología , Tuberculosis/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/microbiología , Humanos , Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Linfocitos T/microbiología , Tuberculosis/microbiología
7.
Front Microbiol ; 10: 2102, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552007

RESUMEN

Tuberculosis remains a public health problem and a main cause of death to humans. Both Mycobacterium tuberculosis and Mycobacterium africanum cause tuberculosis. In contrast to M. tuberculosis, which is geographically spread, M. africanum is restricted to West Africa. Differences have also been found in the growth rate and type of disease caused by M. africanum, globally suggesting an attenuation of this bacteria. In this study, we used the mouse model of infection to follow the dynamics of M. africanum infection in terms of bacterial burdens and tissue pathology, as well as the immune response triggered. Our findings support a lower virulence of M. africanum as compared to M. tuberculosis, including in mice lacking IFN-γ, a major protective cytokine in tuberculosis. Furthermore, the lung immune response triggered by M. africanum infection in wild-type animals was characterized by a discrete influx of leukocytes and a modest transcriptional upregulation of inflammatory mediators. Our findings contribute to elucidate the pathogenesis of M. africanum, supporting the hypothesis that this is an attenuated member of the tuberculosis-causing bacteria. Understanding the biology of M. africanum and how it interacts with the host to establish infection will have implications for our knowledge of TB and for the development of novel and better tools to control this devastating disease.

8.
PLoS Pathog ; 13(8): e1006421, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28817682

RESUMEN

Tuberculosis (TB) is a devastating disease to mankind that has killed more people than any other infectious disease. Despite many efforts and successes from the scientific and health communities, the prospect of TB elimination remains distant. On the one hand, sustainable public health programs with affordable and broad implementation of anti-TB measures are needed. On the other hand, achieving TB elimination requires critical advances in three areas: vaccination, diagnosis, and treatment. It is also well accepted that succeeding in advancing these areas requires a deeper knowledge of host-pathogen interactions during infection, and for that, better experimental models are needed. Here, we review the potential and limitations of different experimental approaches used in TB research, focusing on animal and human-based cell culture models. We highlight the most recent advances in developing in vitro 3D models and introduce the potential of lung organoids as a new tool to study Mycobacterium tuberculosis infection.


Asunto(s)
Modelos Animales de Enfermedad , Modelos Biológicos , Organoides , Tuberculosis , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...