Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
J Antibiot (Tokyo) ; 77(4): 245-256, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38238588

RESUMEN

Tunicamycins (TUN) are well-defined, Streptomyces-derived natural products that inhibit protein N-glycosylation in eukaryotes, and by a conserved mechanism also block bacterial cell wall biosynthesis. TUN inhibits the polyprenylphosphate-N-acetyl-hexosamine-1-phospho-transferases (PNPT), an essential family of enzymes found in both bacteria and eukaryotes. We have previously published the development of chemically modified TUN, called TunR1 and TunR2, that have considerably reduced activity on eukaryotes but that retain the potent antibacterial properties. A mechanism for this reduced toxicity has also been reported. TunR1 and TunR2 have been tested against mammalian cell lines in culture and against live insect cells but, until now, no in vivo evaluation has been undertaken for vertebrates. In the current work, TUN, TunR1, and TunR2 are investigated for their relative toxicity and antimycobacterial activity in zebrafish using a well-established Mycobacterium marinum (M. marinum) infection system, a model for studying human Mycobacterium tuberculosis infections. We also report the relative ability to activate the unfolded protein response (UPR), the known mechanism for the eukaryotic toxicity observed with TUN treatment. Importantly, TunR1 and TunR2 retained their antimicrobial properties, as evidenced by a reduction in M. marinum bacterial burden, compared to DMSO-treated zebrafish. In summary, findings from this study highlight the characteristics of recently developed TUN derivatives, mainly TunR2, and its potential for use as a novel anti-bacterial agent for veterinary and potential medical purposes.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas , Mycobacterium marinum , Tunicamicina , Animales , Humanos , Antibacterianos/farmacología , Mamíferos , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/fisiología , Tunicamicina/química , Tunicamicina/análogos & derivados , Pez Cebra/microbiología , Fosfotransferasas/química
2.
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
3.
Microbiol Mol Biol Rev ; 84(4)2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-32878966

RESUMEN

Pathogenic mycobacteria cause chronic and acute diseases ranging from human tuberculosis (TB) to nontubercular infections. Mycobacterium tuberculosis causes both acute and chronic human tuberculosis. Environmentally acquired nontubercular mycobacteria (NTM) cause chronic disease in humans and animals. Not surprisingly, NTM and M. tuberculosis often use shared molecular mechanisms to survive within the host. The ESX-1 system is a specialized secretion system that is essential for virulence and is functionally conserved between M. tuberculosis and Mycobacterium marinumM. marinum is an NTM found in both salt water and freshwater that is often used to study mycobacterial virulence. Since the discovery of the secretion system in 2003, the use of both M. tuberculosis and M. marinum has defined the conserved molecular mechanisms underlying protein secretion and the lytic and regulatory activities of the ESX-1 system. Here, we review the trajectory of the field, including key discoveries regarding the ESX-1 system. We highlight the contributions of M. marinum studies and the conserved and unique aspects of the ESX-1 secretion system.


Asunto(s)
Mycobacterium marinum/fisiología , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/fisiología , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Sistemas de Secreción Tipo VII/fisiología , Animales , Proteínas Bacterianas/fisiología , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Modelos Biológicos , Transporte de Proteínas , Virulencia
4.
PLoS One ; 15(7): e0233252, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32701962

RESUMEN

Phthiocerol dimycocerosates (PDIMs) are a class of mycobacterial lipids that promote virulence in Mycobacterium tuberculosis and Mycobacterium marinum. It has recently been shown that PDIMs work in concert with the M. tuberculosis Type VII secretion system ESX-1 to permeabilize the phagosomal membranes of infected macrophages. As the zebrafish-M. marinum model of infection has revealed the critical role of PDIM at the host-pathogen interface, we set to determine if PDIMs contributed to phagosomal permeabilization in M. marinum. Using an ΔmmpL7 mutant defective in PDIM transport, we find the PDIM-ESX-1 interaction to be conserved in an M. marinum macrophage infection model. However, we find PDIM and ESX-1 mutants differ in their degree of defect, with the PDIM mutant retaining more membrane damaging activity. Using an in vitro hemolysis assay-a common surrogate for cytolytic activity, we find that PDIM and ESX-1 differ in their contributions: the ESX-1 mutant loses hemolytic activity while PDIM retains it. Our observations confirm the involvement of PDIMs in phagosomal permeabilization in M. marinum infection and suggest that PDIM enhances the membrane disrupting activity of pathogenic mycobacteria and indicates that the role they play in damaging phagosomal and red blood cell membranes may differ.


Asunto(s)
Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Lípidos/farmacología , Macrófagos/citología , Mycobacterium marinum/metabolismo , Fagosomas/efectos de los fármacos , Línea Celular , Humanos , Macrófagos/efectos de los fármacos , Mycobacterium marinum/fisiología , Permeabilidad/efectos de los fármacos , Fagosomas/metabolismo
5.
FEBS J ; 287(18): 3925-3943, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32485057

RESUMEN

Multi-drug-resistant tuberculosis is a worldwide problem, and there is an urgent need for host-derived therapeutic targets, circumventing emerging drug resistance. We have previously shown that hypoxia-inducible factor-1α (Hif-1α) stabilisation helps the host to clear mycobacterial infection via neutrophil activation. However, Hif-1α stabilisation has also been implicated in chronic inflammatory diseases caused by prolonged neutrophilic inflammation. Comorbid infection and inflammation can be found together in disease settings, and it remains unclear whether Hif-1α stabilisation would be beneficial in a holistic disease setting. Here, we set out to understand the effects of Hif-1α on neutrophil behaviour in a comorbid setting by combining two well-characterised in vivo zebrafish models - TB infection (Mycobacterium marinum infection) and sterile injury (tailfin transection). Using a local Mm infection near to the tailfin wound site caused neutrophil migration between the two sites that was reduced during Hif-1α stabilisation. During systemic Mm infection, wounding leads to increased infection burden, but the protective effect of Hif-1α stabilisation remains. Our data indicate that Hif-1α stabilisation alters neutrophil migration dynamics between comorbid sites and that the protective effect of Hif-1α against Mm is maintained in the presence of inflammation, highlighting its potential as a host-derived target against TB infection.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/inmunología , Neutrófilos/inmunología , Proteínas de Pez Cebra/inmunología , Pez Cebra/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Larva/inmunología , Larva/metabolismo , Larva/microbiología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/fisiología , Neutrófilos/metabolismo , Estabilidad Proteica , Pez Cebra/metabolismo , Pez Cebra/microbiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
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
7.
Curr Opin Microbiol ; 54: 103-110, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32062573

RESUMEN

Thirty years ago Stanley Falkow formulated molecular Koch's postulates as a framework to help dissect the contribution of microbial genes to their pathogenicity (Box 1). Three years later, his advice led me to develop Mycobacterium marinum, a close genetic relative of Mycobacterium tuberculosis, as a model for tuberculosis pathogenesis. Here, I discuss insights into M. tuberculosis pathogenicity from studying M. marinum in the zebrafish, and frame them in terms of molecular Koch's postulates. The highly orchestrated life cycle of M. tuberculosis is achieved in substantial measure not by "traditional" pathogen-exclusive virulence genes acquired along its evolutionary history, but rather by genes that are shared with its environmental ancestors. Together, these genes support its tactics of subterfuge and exploitation to overcome host immunity so as to produce the transmissible disease that ensures the evolutionary survival of this obligate human pathogen.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Factores de Virulencia/fisiología , Animales , Modelos Animales de Enfermedad , Genes Bacterianos , Interacciones Huésped-Patógeno , Humanos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/fisiología , Mycobacterium tuberculosis/fisiología , Tuberculosis/inmunología , Virulencia/genética , Factores de Virulencia/genética , Pez Cebra
8.
J Leukoc Biol ; 107(2): 185-203, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31529512

RESUMEN

The CXCR3-CXCL11 chemokine-signaling axis plays an essential role in infection and inflammation by orchestrating leukocyte trafficking in human and animal models, including zebrafish. Atypical chemokine receptors (ACKRs) play a fundamental regulatory function in signaling networks by shaping chemokine gradients through their ligand scavenging function, while being unable to signal in the classic G-protein-dependent manner. Two copies of the CXCR3 gene in zebrafish, cxcr3.2 and cxcr3.3, are expressed on macrophages and share a highly conserved ligand-binding site. However, Cxcr3.3 has structural characteristics of ACKRs indicative of a ligand-scavenging role. In contrast, we previously showed that Cxcr3.2 is an active CXCR3 receptor because it is required for macrophage motility and recruitment to sites of mycobacterial infection. In this study, we generated a cxcr3.3 CRISPR-mutant to functionally dissect the antagonistic interplay among the cxcr3 paralogs in the immune response. We observed that cxcr3.3 mutants are more susceptible to mycobacterial infection, whereas cxcr3.2 mutants are more resistant. Furthermore, macrophages in the cxcr3.3 mutant are more motile, show higher activation status, and are recruited more efficiently to sites of infection or injury. Our results suggest that Cxcr3.3 is an ACKR that regulates the activity of Cxcr3.2 by scavenging common ligands and that silencing the scavenging function of Cxcr3.3 results in an exacerbated Cxcr3.2 signaling. In human, splice variants of CXCR3 have antagonistic functions and CXCR3 ligands also interact with ACKRs. Therefore, in zebrafish, an analogous regulatory mechanism appears to have evolved after the cxcr3 gene duplication event, through diversification of conventional and atypical receptor variants.


Asunto(s)
Movimiento Celular , Macrófagos/fisiología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/fisiología , Receptores CXCR3/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Sistemas CRISPR-Cas , Macrófagos/citología , Macrófagos/microbiología , Mutación , Infecciones por Mycobacterium no Tuberculosas/metabolismo , Infecciones por Mycobacterium no Tuberculosas/patología , Conformación Proteica , Receptores CXCR3/antagonistas & inhibidores , Receptores CXCR3/clasificación , Receptores CXCR3/genética , Pez Cebra/microbiología , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
9.
Fish Shellfish Immunol ; 96: 78-85, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31775059

RESUMEN

Macrophages are the first-line host defense that the invading Mycobacterium tuberculosis (Mtb) encounters. It has been recently reported that host aerobic glycolysis was elevated post the infection by a couple of virulent mycobacterial species. However, whether this metabolic transition is required for host defense against intracellular pathogens and the underlying mechanisms remain to be further investigated. A pathogenic mycobacterial species, M. marinum, is genetically close to Mtb and was utilized in this study. Through analyzing cellular carbon metabolism of RAW 264.7 (a murine macrophage-like cell line) post M. marinum infection, a strong elevation of glycolysis was observed. Next, three glycolysis inhibitors were examined for their ability to inhibit mycobacterial proliferation inside RAW264.7 macrophages. Among them, a glucose analog, 2-deoxyglucose (2-DG) displayed a protective role against mycobacterial infection. Treatment with 2-DG at concentrations of 0.5 or 1 mM significantly induced autophagy and decreased the phagocytosis of M. marinum by macrophages. Moreover, 2-DG pre-treatment exerted a significantly protective effect on zebrafish larvae by limiting the proliferation of M. marinum, and such effect was correlated to tumor necrosis factor alpha (TNF-α) as the 2-DG pre-treatment increased the expression of TNF-α in both mouse peritoneal macrophages and zebrafish. On the contrary, the 2-DG treatment post infection did not restrain proliferation of M. marinum in WT zebrafish, and even accelerated bacterial replication in TNF-α-/- zebrafish. Together, modulation of glycolysis prior to infection boosts host immunity against M. marinum infection, indicating a potential intervention strategy to control mycobacterial infection.


Asunto(s)
Enfermedades de los Peces/metabolismo , Glucólisis , Infecciones por Mycobacterium no Tuberculosas/veterinaria , Mycobacterium marinum/fisiología , Pez Cebra , Aerobiosis , Animales , Enfermedades de los Peces/microbiología , Ratones , Infecciones por Mycobacterium no Tuberculosas/metabolismo , Infecciones por Mycobacterium no Tuberculosas/microbiología , Células RAW 264.7
10.
BMC Genomics ; 20(1): 961, 2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31823727

RESUMEN

BACKGROUND: During infection by intracellular pathogens, a highly complex interplay occurs between the infected cell trying to degrade the invader and the pathogen which actively manipulates the host cell to enable survival and proliferation. Many intracellular pathogens pose important threats to human health and major efforts have been undertaken to better understand the host-pathogen interactions that eventually determine the outcome of the infection. Over the last decades, the unicellular eukaryote Dictyostelium discoideum has become an established infection model, serving as a surrogate macrophage that can be infected with a wide range of intracellular pathogens. In this study, we use high-throughput RNA-sequencing to analyze the transcriptional response of D. discoideum when infected with Mycobacterium marinum and Legionella pneumophila. The results were compared to available data from human macrophages. RESULTS: The majority of the transcriptional regulation triggered by the two pathogens was found to be unique for each bacterial challenge. Hallmark transcriptional signatures were identified for each infection, e.g. induction of endosomal sorting complexes required for transport (ESCRT) and autophagy genes in response to M. marinum and inhibition of genes associated with the translation machinery and energy metabolism in response to L. pneumophila. However, a common response to the pathogenic bacteria was also identified, which was not induced by non-pathogenic food bacteria. Finally, comparison with available data sets of regulation in human monocyte derived macrophages shows that the elicited response in D. discoideum is in many aspects similar to what has been observed in human immune cells in response to Mycobacterium tuberculosis and L. pneumophila. CONCLUSIONS: Our study presents high-throughput characterization of D. discoideum transcriptional response to intracellular pathogens using RNA-seq. We demonstrate that the transcriptional response is in essence distinct to each pathogen and that in many cases, the corresponding regulation is recapitulated in human macrophages after infection by mycobacteria and L. pneumophila. This indicates that host-pathogen interactions are evolutionary conserved, derived from the early interactions between free-living phagocytic cells and bacteria. Taken together, our results strengthen the use of D. discoideum as a general infection model.


Asunto(s)
Infecciones Bacterianas/microbiología , Dictyostelium/microbiología , Modelos Biológicos , Proteínas Protozoarias/genética , Células Cultivadas , Citoplasma/microbiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Legionella pneumophila/fisiología , Macrófagos/microbiología , Mycobacterium marinum/fisiología , Proteínas Protozoarias/metabolismo , Especificidad de la Especie , Transcripción Genética
11.
J Fish Dis ; 42(10): 1425-1431, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31418901

RESUMEN

The zebrafish (Danio rerio) is a popular vertebrate model organism used in a wide range of research fields. Mycobacteriosis, caused by Mycobacterium species, is particularly concerning because it is a common disease associated with chronic infections in these fish. Infections are also a source of uncontrolled experimental variance that may influence research results. Live feeds for zebrafish are common and include paramecia (Paramecium caudatum), brine shrimp (Artemia franciscana) and rotifers (Branchionus spp.). Although nutritionally beneficial, live feeds may pose a biosecurity risk. In this study, we investigate transmission of Mycobacterium chelonae and Mycobacterium marinum through these three live feeds. We show that all three live feeds ingest both M. marinum and M. chelonae and can transmit mycobacterial infections to zebrafish. This observation emphasizes the need for live feeds to be included in the consideration of potential biosecurity risks. This study is of importance to other beyond the zebrafish community, including those of additional aquatic models and those using live feeds for other types of aquaculture.


Asunto(s)
Alimentación Animal/microbiología , Enfermedades de los Peces/transmisión , Infecciones por Mycobacterium no Tuberculosas/veterinaria , Mycobacterium chelonae/fisiología , Mycobacterium marinum/fisiología , Pez Cebra , Animales , Artemia/microbiología , Dieta/veterinaria , Femenino , Enfermedades de los Peces/epidemiología , Enfermedades de los Peces/microbiología , Masculino , Infecciones por Mycobacterium no Tuberculosas/epidemiología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Infecciones por Mycobacterium no Tuberculosas/transmisión , Paramecium caudatum/microbiología , Prevalencia , Rotíferos/microbiología
12.
Sci Rep ; 9(1): 995, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30700796

RESUMEN

Tuberculosis is a multifactorial bacterial disease, which can be modeled in the zebrafish (Danio rerio). Abdominal cavity infection with Mycobacterium marinum, a close relative of Mycobacterium tuberculosis, leads to a granulomatous disease in adult zebrafish, which replicates the different phases of human tuberculosis, including primary infection, latency and spontaneous reactivation. Here, we have carried out a transcriptional analysis of zebrafish challenged with low-dose of M. marinum, and identified intelectin 3 (itln3) among the highly up-regulated genes. In order to clarify the in vivo significance of Itln3 in immunity, we created nonsense itln3 mutant zebrafish by CRISPR/Cas9 mutagenesis and analyzed the outcome of M. marinum infection in both zebrafish embryos and adult fish. The lack of functional itln3 did not affect survival or the mycobacterial burden in the zebrafish. Furthermore, embryonic survival was not affected when another mycobacterial challenge responsive intelectin, itln1, was silenced using morpholinos either in the WT or itln3 mutant fish. In addition, M. marinum infection in dexamethasone-treated adult zebrafish, which have lowered lymphocyte counts, resulted in similar bacterial burden in both WT fish and homozygous itln3 mutants. Collectively, although itln3 expression is induced upon M. marinum infection in zebrafish, it is dispensable for protective mycobacterial immune response.


Asunto(s)
Citocinas/metabolismo , Lectinas/metabolismo , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/microbiología , Animales , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Codón sin Sentido/genética , Citocinas/genética , Dexametasona/farmacología , Resistencia a la Enfermedad/inmunología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Embrión no Mamífero/microbiología , Regulación de la Expresión Génica/efectos de los fármacos , Genoma , Lectinas/genética , Depleción Linfocítica , Morfolinos/farmacología , Mutación/genética , Infecciones por Mycobacterium no Tuberculosas/genética , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/efectos de los fármacos , Análisis de Supervivencia , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
13.
Dev Comp Immunol ; 92: 253-259, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30521838

RESUMEN

Owing to the high incidence of multi-drug resistance and challenges posed by the complex and long duration of treatments, Mycobacterium tuberculosis (Mtb) infections remain a significant clinical burden, which would benefit from development of novel immuno-therapeutic-based treatment strategies. Among early immune effectors, invariant or innate-like (i)T cells are attracting attention because of their potential regulatory activity, which can shape anti-mycobacterial immune responses. Unlike conventional T cells, iT cells express a semi-invariant T cell receptor, and respond rapidly and robustly to molecular patterns presented by MHC class I-like molecules. To date, functional studies of iT cells in vivo has been problematic and the role of iT cells in anti-Mtb responses remains unclear. Here, after reviewing the recent literature on anti-mycobacterial iT cell immunity, we describe a novel alternative model system in the amphibian Xenopus laevis tadpoles during infection with Mycobacterium marinum (Mm). X. laevis tadpoles rely mostly on a few distinct prominent innate-like (i)T cell subsets, whose development and function are governed by distinct MHC class I-like molecules. Thus, X. laevis tadpoles provide a convenient and cost-effective in vivo model uniquely suited to investigate the roles of iT cells during mycobacterial infections. We have developed reverse genetics and MHC tetramer technology to characterize this MHC-like/iT system in tadpoles. Our study in X. laevis provides evidence of a conserved convergent function of iT cells in host defenses against mycobacteria between mammals and amphibians.


Asunto(s)
Mycobacterium marinum/fisiología , Mycobacterium tuberculosis/fisiología , Células T Asesinas Naturales/inmunología , Linfocitos T/inmunología , Tuberculosis/inmunología , Xenopus/inmunología , Animales , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunidad Celular , Inmunidad Innata , Modelos Animales
14.
J Immunol ; 202(2): 494-502, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30552162

RESUMEN

Drug-resistant mycobacteria are a rising problem worldwide. There is an urgent need to understand the immune response to tuberculosis to identify host targets that, if targeted therapeutically, could be used to tackle these currently untreatable infections. In this study we use an Il-1ß fluorescent transgenic line to show that there is an early innate immune proinflammatory response to well-established zebrafish models of inflammation and Mycobacterium marinum infection. We demonstrate that host-derived hypoxia signaling, mediated by the Hif-1α transcription factor, can prime macrophages with increased levels of Il-1ß in the absence of infection, upregulating neutrophil antimicrobial NO production, leading to greater protection against infection. Our data link Hif-1α to proinflammatory macrophage Il-1ß transcription in vivo during early mycobacterial infection and importantly highlight a host protective mechanism, via antimicrobial NO, that decreases disease outcomes and that could be targeted therapeutically to stimulate the innate immune response to better deal with infections.


Asunto(s)
Hipoxia/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/fisiología , Mycobacterium tuberculosis/fisiología , Tuberculosis/inmunología , Animales , Animales Modificados Genéticamente , Antituberculosos/metabolismo , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Proteínas Fluorescentes Verdes/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunidad Innata , Interleucina-1beta/genética , Óxido Nítrico/metabolismo , Pez Cebra
15.
J Vis Exp ; (143)2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31929505

RESUMEN

Zebrafish is an excellent model organism for studying innate immune cell behavior due to its transparent nature and reliance solely on its innate immune system during early development. The Zebrafish Mycobacterium marinum (M. marinum) infection model has been well-established in studying host immune response against mycobacterial infection. It has been suggested that different macrophage cell death types will lead to the diverse outcomes of mycobacterial infection. Here we describe a protocol using intravital microscopy to observe macrophage cell death in zebrafish embryos following M. marinum infection. Zebrafish transgenic lines that specifically label macrophages and neutrophils are infected via intramuscular microinjection of fluorescently labeled M. marinum in either the midbrain or the trunk. Infected zebrafish embryos are subsequently mounted on low melting agarose and observed by confocal microscopy in X-Y-Z-T dimensions. Because long-term live imaging requires using low laser power to avoid photobleaching and phototoxicity, a strongly expressing transgenic is highly recommended. This protocol facilitates the visualization of the dynamic processes in vivo, including immune cell migration, host pathogen interaction, and cell death.


Asunto(s)
Muerte Celular , Embrión no Mamífero/inmunología , Embrión no Mamífero/microbiología , Macrófagos/citología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/fisiología , Pez Cebra/embriología , Animales , Movimiento Celular , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Macrófagos/inmunología , Neutrófilos/citología
16.
J Vis Exp ; (140)2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30346391

RESUMEN

Mycobacterium tuberculosis is currently the deadliest human pathogen causing 1.7 million deaths and 10.4 million infections every year. Exposure to this bacterium causes a wide disease spectrum in humans ranging from a sterilized infection to an actively progressing deadly disease. The most common form is the latent tuberculosis, which is asymptomatic, but has the potential to reactivate into a fulminant disease. Adult zebrafish and its natural pathogen Mycobacterium marinum have recently proven to be an applicable model to study the wide disease spectrum of tuberculosis. Importantly, spontaneous latency and reactivation as well as adaptive immune responses in the context of mycobacterial infection can be studied in this model. In this article, we describe methods for the experimental infection of adult zebrafish, the collection of internal organs for the extraction of nucleic acids for the measurement of mycobacterial loads and host immune responses by quantitative PCR. The in-house-developed, M. marinum-specific qPCR assay is more sensitive than the traditional plating methods as it also detects DNA from non-dividing, dormant or recently dead mycobacteria. As both DNA and RNA are extracted from the same individual, it is possible to study the relationships between the diseased state, and the host and pathogen gene-expression. The adult zebrafish model for tuberculosis thus presents itself as a highly applicable, non-mammalian in vivo system to study host-pathogen interactions.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Mycobacterium no Tuberculosas/inmunología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/fisiología , Pez Cebra , Animales , Expresión Génica , Interacciones Huésped-Patógeno , Infecciones por Mycobacterium no Tuberculosas/genética , Mycobacterium marinum/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
J Cell Sci ; 131(17)2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30054386

RESUMEN

Phagocytic cells take up, kill and digest microbes by a process called phagocytosis. To this end, these cells bind the particle, rearrange their actin cytoskeleton, and orchestrate transport of digestive factors to the particle-containing phagosome. The mammalian lysosomal membrane protein LIMP-2 (also known as SCARB2) and CD36, members of the class B of scavenger receptors, play a crucial role in lysosomal enzyme trafficking and uptake of mycobacteria, respectively, and generally in host cell defences against intracellular pathogens. Here, we show that the Dictyostelium discoideum LIMP-2 homologue LmpA regulates phagocytosis and phagolysosome biogenesis. The lmpA knockdown mutant is highly affected in actin-dependent processes, such as particle uptake, cellular spreading and motility. Additionally, the cells are severely impaired in phagosomal acidification and proteolysis, likely explaining the higher susceptibility to infection with the pathogenic bacterium Mycobacterium marinum, a close cousin of the human pathogen Mycobacterium tuberculosis Furthermore, we bring evidence that LmpB is a functional homologue of CD36 and specifically mediates uptake of mycobacteria. Altogether, these data indicate a role for LmpA and LmpB, ancestors of the family of which LIMP-2 and CD36 are members, in lysosome biogenesis and host cell defence.


Asunto(s)
Dictyostelium/fisiología , Proteínas de Membrana de los Lisosomas/metabolismo , Mycobacterium marinum/fisiología , Fagocitosis , Proteínas Protozoarias/metabolismo , Receptores de Lipoproteína/metabolismo , Antígenos CD36/genética , Dictyostelium/genética , Dictyostelium/microbiología , Humanos , Proteínas de Membrana de los Lisosomas/genética , Proteínas Protozoarias/genética , Receptores de Lipoproteína/genética , Receptores Depuradores/genética
18.
Crit Rev Microbiol ; 44(6): 779-792, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30663918

RESUMEN

Tuberculosis is still a global health burden. It is caused by Mycobacterium tuberculosis which afflicts around one third of the world's population and costs around 1.3 million people their lives every year. Bacillus Calmette-Guerin vaccine is inefficient to prevent overt infection. Additionally, the lengthy inconvenient course of treatment, along with the raising issue of antimicrobial resistance, result in incomplete eradication of this infectious disease. The lack of proper animal models that replicate the latent and active courses of human tuberculosis infection remains one of the main reasons behind the poor advancement in tuberculosis research. Danio rerio, commonly known as zebrafish, is catching more attention as an animal model in tuberculosis research field. This shift is based on the histological and pathological similarities between Mycobacterium marinum infection in zebrafish and Mycobacterium tuberculosis infection in humans. Being small, cheap, transparent, and easy to handle have added further advantages to the use of zebrafish model. Besides better understanding of the pathogenesis of tuberculosis, Mycobacterium marinum infected zebrafish model is useful for evaluating novel vaccines against human tuberculosis, high throughput small molecule screening, repurposing established drugs with possible antitubercular activity, and assessing novel antituberculars for hepatotoxicity.


Asunto(s)
Modelos Animales de Enfermedad , Tuberculosis/microbiología , Pez Cebra , Animales , Antituberculosos/farmacología , Humanos , Mycobacterium marinum/efectos de los fármacos , Mycobacterium marinum/fisiología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/fisiología , Tuberculosis/tratamiento farmacológico , Pez Cebra/microbiología
19.
Microb Cell Fact ; 16(1): 217, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-29183333

RESUMEN

BACKGROUND: Although mycobacterial glycolipids are among the first-line molecules involved in host-pathogen interactions, their contribution in virulence remains incomplete. Mycobacterium marinum is a waterborne pathogen of fish and other ectotherms, closely related to Mycobacterium tuberculosis. Since it causes tuberculosis-like systemic infection it is widely used as a model organism for studying the pathogenesis of tuberculosis. It is also an occasional opportunistic human pathogen. The M. marinum surface-exposed lipooligosaccharides (LOS) are immunogenic molecules that participate in the early interactions with macrophages and modulate the host immune system. Four major LOS species, designated LOS-I to LOS-IV, have been identified and characterized in M. marinum. Herein, we investigated the interactions between a panel of defined M. marinum LOS mutants that exhibited various degrees of truncation in the LOS structure, and human-derived THP-1 macrophages to address the potential of LOSs to act as pro- or avirulence factors. RESULTS: A moderately truncated LOS structure did not interfere with M. marinum invasion. However, a deeper shortening of the LOS structure was associated with increased entry of M. marinum into host cells and increased elimination of the bacilli by the macrophages. These effects were dependent on Toll-like receptor 2. CONCLUSION: We provide the first evidence that LOSs inhibit the interaction between mycobacterial cell wall ligands and appropriate macrophage pattern recognition receptors, affecting uptake and elimination of the bacteria by host phagocytes.


Asunto(s)
Lipopolisacáridos/genética , Lipopolisacáridos/inmunología , Macrófagos/microbiología , Mycobacterium marinum/inmunología , Receptor Toll-Like 2/inmunología , Línea Celular , Pared Celular/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/biosíntesis , Macrófagos/inmunología , Mycobacterium marinum/química , Mycobacterium marinum/patogenicidad , Mycobacterium marinum/fisiología , Factores de Virulencia
20.
PLoS One ; 12(7): e0181121, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28727774

RESUMEN

Tuberculosis remains one of the major threats to public health worldwide. Given the prevalence of multi drug resistance (MDR) in Mycobacterium tuberculosis strains, there is a strong need to develop new anti-mycobacterial drugs with modes of action distinct from classical antibiotics. Inhibitors of mycobacterial virulence might target new molecular processes and may represent a potential new therapeutic alternative. In this study, we used a Dictyostelium discoideum host model to assess virulence of Mycobacterium marinum and to identify compounds inhibiting mycobacterial virulence. Among 9995 chemical compounds, we selected 12 inhibitors of mycobacterial virulence that do not inhibit mycobacterial growth in synthetic medium. Further analyses revealed that 8 of them perturbed functions requiring an intact mycobacterial cell wall such as sliding motility, bacterial aggregation or cell wall permeability. Chemical analogs of two compounds were analyzed. Chemical modifications altered concomitantly their effect on sliding motility and on mycobacterial virulence, suggesting that the alteration of the mycobacterial cell wall caused the loss of virulence. We characterized further one of the selected compounds and found that it inhibited the ability of mycobacteria to replicate in infected cells. Together these results identify new antimycobacterial compounds that represent new tools to unravel the molecular mechanisms controlling mycobacterial pathogenicity. The isolation of compounds with anti-virulence activity is the first step towards developing new antibacterial treatments.


Asunto(s)
Dictyostelium/microbiología , Mycobacterium marinum/efectos de los fármacos , Virulencia/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Mycobacterium marinum/patogenicidad , Mycobacterium marinum/fisiología , Mycobacterium marinum/ultraestructura , Bibliotecas de Moléculas Pequeñas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...