Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
mSphere ; 6(3)2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33952660

RESUMEN

Mycobacterium tuberculosis infections claim more than a million lives each year, and better treatments or vaccines are required. A crucial pathogenicity factor is translocation from phagolysosomes to the cytosol upon phagocytosis by macrophages. Translocation from the phagolysosome to the cytosol is an ESX-1-dependent process, as previously shown in vitro Here, we show that in vivo, mycobacteria also translocate to the cytosol but mainly when host immunity is compromised. We observed only low numbers of cytosolic bacilli in mice, armadillos, zebrafish, and patient material infected with M. tuberculosis, M. marinum, or M. leprae In contrast, when innate or adaptive immunity was compromised, as in severe combined immunodeficiency (SCID) or interleukin-1 receptor 1 (IL-1R1)-deficient mice, significant numbers of cytosolic M. tuberculosis bacilli were detected in the lungs of infected mice. Taken together, in vivo, translocation to the cytosol of M. tuberculosis is controlled by adaptive immune responses as well as IL-1R1-mediated signals.IMPORTANCE For decades, Mycobacterium tuberculosis has been one of the deadliest pathogens known. Despite infecting approximately one-third of the human population, no effective treatment or vaccine is available. A crucial pathogenicity factor is subcellular localization, as M. tuberculosis can translocate from phagolysosome to the cytosol in macrophages. The situation in vivo is more complicated. In this study, we establish that high-level cytosolic escape of mycobacteria can indeed occur in vivo but mainly when host resistance is compromised. The IL-1 pathway is crucial for the control of the number of cytosolic mycobacteria. The establishment that immune signals result in the clearance of cells containing cytosolic mycobacteria connects two important fields, cell biology and immunology, which is vital for the understanding of the pathology of M. tuberculosis.


Asunto(s)
Citosol/microbiología , Mycobacterium/inmunología , Mycobacterium/patogenicidad , Fagosomas/microbiología , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Transducción de Señal/inmunología , Animales , Armadillos/microbiología , Traslocación Bacteriana , Citosol/inmunología , Femenino , Humanos , Lepra/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Mycobacterium/clasificación , Fagosomas/inmunología , Piel/microbiología , Piel/patología , Células THP-1 , Pez Cebra
2.
Am J Hum Genet ; 107(5): 989-999, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33053334

RESUMEN

Osteogenesis imperfecta (OI) is characterized primarily by susceptibility to fractures with or without bone deformation. OI is genetically heterogeneous: over 20 genetic causes are recognized. We identified bi-allelic pathogenic KDELR2 variants as a cause of OI in four families. KDELR2 encodes KDEL endoplasmic reticulum protein retention receptor 2, which recycles ER-resident proteins with a KDEL-like peptide from the cis-Golgi to the ER through COPI retrograde transport. Analysis of patient primary fibroblasts showed intracellular decrease of HSP47 and FKBP65 along with reduced procollagen type I in culture media. Electron microscopy identified an abnormal quality of secreted collagen fibrils with increased amount of HSP47 bound to monomeric and multimeric collagen molecules. Mapping the identified KDELR2 variants onto the crystal structure of G. gallus KDELR2 indicated that these lead to an inactive receptor resulting in impaired KDELR2-mediated Golgi-ER transport. Therefore, in KDELR2-deficient individuals, OI most likely occurs because of the inability of HSP47 to bind KDELR2 and dissociate from collagen type I. Instead, HSP47 remains bound to collagen molecules extracellularly, disrupting fiber formation. This highlights the importance of intracellular recycling of ER-resident molecular chaperones for collagen type I and bone metabolism and a crucial role of HSP47 in the KDELR2-associated pathogenic mechanism leading to OI.


Asunto(s)
Huesos/metabolismo , Colágeno Tipo I/metabolismo , Proteínas del Choque Térmico HSP47/metabolismo , Osteogénesis Imperfecta/genética , Proteínas de Transporte Vesicular/metabolismo , Adulto , Alelos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Huesos/patología , Pollos , Preescolar , Colágeno Tipo I/química , Colágeno Tipo I/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Expresión Génica , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Proteínas del Choque Térmico HSP47/química , Proteínas del Choque Térmico HSP47/genética , Humanos , Lactante , Masculino , Osteogénesis Imperfecta/diagnóstico , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología , Linaje , Cultivo Primario de Células , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Transporte de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética
3.
Clin Transl Sci ; 13(6): 1060-1064, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32267997

RESUMEN

The zebrafish infected with Mycobacterium marinum (M. marinum) is an attractive tuberculosis disease model, showing similar pathogenesis to Mycobacterium tuberculosis (M. tuberculosis) infections in humans. To translate pharmacological findings from this disease model to higher vertebrates, a quantitative understanding of the natural growth of M. marinum in comparison to the natural growth of M. tuberculosis is essential. Here, the natural growth of two strains of M. marinum, E11 and MUSA , is studied over an extended period using an established model-based approach, the multistate tuberculosis pharmacometric (MTP) model, for comparison to that of M. tuberculosis. Poikilotherm-derived strain E11 and human-derived strain MUSA were grown undisturbed up to 221 days and viability of cultures (colony forming unit (CFU)/mL) was determined by plating at different time points. Nonlinear mixed effects modeling using the MTP model quantified the bacterial growth, the transfer among fast, slow, and non-multiplying states, and the inoculi. Both strains showed initial logistic growth, reaching a maximum after 20-25 days for E11 and MUSA , respectively, followed by a decrease to a new plateau. Natural growth of both E11 and MUSA was best described with Gompertz growth functions. For E11, the inoculum was best described in the slow-multiplying state, for MUSA in the fast-multiplying state. Natural growth of E11 was most similar to that of M. tuberculosis, whereas MUSA showed more aggressive growth behavior. Characterization of natural growth of M. marinum and quantitative comparison with M. tuberculosis brings the zebrafish tuberculosis disease model closer to the quantitative translational pipeline of antituberculosis drug development.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium marinum/crecimiento & desarrollo , Tuberculosis/tratamiento farmacológico , Animales , Antituberculosos/uso terapéutico , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Humanos , Modelos Biológicos , Mycobacterium marinum/efectos de los fármacos , Mycobacterium marinum/aislamiento & purificación , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Pez Cebra/microbiología
4.
mBio ; 10(5)2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31662454

RESUMEN

Tuberculosis, one of the world's most severe infectious diseases, is caused by Mycobacterium tuberculosis A major weapon of this pathogen is a unique cell wall that protects the pathogen from eradication by the immune system. Mycobacteria have specialized secretion systems, e.g., type VII secretion or ESX systems, to transport substrates across this cell wall. The largest group of proteins that are secreted by these ESX systems are the PE proteins. Previously, it was shown that the N-terminal PE domain of about 100 amino acids is required for secretion. Here, we describe the identification of an aspartic protease, designated PecA, that removes (part of) this PE domain at the cell surface. Nearly all of the observed PE_PGRS proteins are processed by PecA. Interestingly, the protease itself is also a secreted PE protein and subject to self-cleavage. Furthermore, a defect in surface processing has no effect on the activity of the PE lipase protein LipY but does seem to affect the functioning of other virulence factors, as a pecA mutant strain of Mycobacterium marinum shows moderate attenuation in zebrafish larvae. In conclusion, our results reveal the presence of a functional aspartic acid protease in M. marinum that cleaves LipY, itself as well as other members of the PE_PGRS family. Finally, mutants lacking PecA show growth attenuation in vivo, suggesting that PecA plays a role during infection.IMPORTANCE Aspartic proteases are common in eukaryotes and retroviruses but are relatively rare among bacteria (N. D. Rawlings and A. Bateman, BMC Genomics 10:437, 2009, https://doi.org/10.1186/1471-2164-10-437). In contrast to eukaryotic aspartic proteases, bacterial aspartic proteases are generally located in the cytoplasm. We have identified a surface-associated mycobacterial aspartic protease, PecA, which cleaves itself and many other type VII secretion substrates of the PE_PGRS family. PecA is present in most pathogenic mycobacterial species, including M. tuberculosis In addition, pathogenicity of M. marinum is reduced in the ΔpecA mutant, indicating that PecA contributes to virulence.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Péptido Hidrolasas/metabolismo , Sistemas de Secreción Tipo VII/metabolismo , Animales , Hidrolasas de Éster Carboxílico , Pared Celular/metabolismo , Larva , Mycobacterium marinum , Virulencia , Factores de Virulencia/metabolismo , Pez Cebra
5.
Sci Rep ; 9(1): 8131, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31148579

RESUMEN

RhoGTPases regulate cytoskeletal dynamics, migration and cell-cell adhesion in endothelial cells. Besides regulation at the level of guanine nucleotide binding, they also undergo post-translational modifications, for example ubiquitination. RhoGTPases are ubiquitinated by Cullin RING ligases which are in turn regulated by neddylation. Previously we showed that inhibition of Cullin RING ligase activity by the neddylation inhibitor MLN4924 is detrimental for endothelial barrier function, due to accumulation of RhoB and the consequent induction of contractility. Here we analyzed the effect of pharmacological activation of Cullin RING ligases on endothelial barrier integrity in vitro and in vivo. CSN5i-3 induced endothelial barrier disruption and increased macromolecule leakage in vitro and in vivo. Mechanistically, CSN5i-3 strongly induced the expression and activation of RhoB and to lesser extent of RhoA in endothelial cells, which enhanced cell contraction. Elevated expression of RhoGTPases was a consequence of activation of the NF-κB pathway. In line with this notion, CSN5i-3 treatment decreased IκBα expression and increased NF-κB-mediated ICAM-1 expression and consequent adhesion of neutrophils to endothelial cells. This study shows that sustained neddylation of Cullin RING-ligases leads to activation the NF-κB pathway in endothelial cells, elevated expression of RhoGTPases, Rho/ROCK-dependent activation of MLC and disruption of the endothelial barrier.


Asunto(s)
Complejo del Señalosoma COP9/metabolismo , Endotelio Vascular/metabolismo , Inflamación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptido Hidrolasas/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoB/metabolismo , Animales , Ciclopentanos/farmacología , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Neutrófilos/metabolismo , Pirimidinas/farmacología , Ubiquitina/química , Regulación hacia Arriba , Pez Cebra
6.
PLoS Pathog ; 14(8): e1007247, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30102741

RESUMEN

The pathogen Mycobacterium tuberculosis employs a range of ESX-1 substrates to manipulate the host and build a successful infection. Although the importance of ESX-1 secretion in virulence is well established, the characterization of its individual components and the role of individual substrates is far from complete. Here, we describe the functional characterization of the Mycobacterium marinum accessory ESX-1 proteins EccA1, EspG1 and EspH, i.e. proteins that are neither substrates nor structural components. Proteomic analysis revealed that EspG1 is crucial for ESX-1 secretion, since all detectable ESX-1 substrates were absent from the cell surface and culture supernatant in an espG1 mutant. Deletion of eccA1 resulted in minor secretion defects, but interestingly, the severity of these secretion defects was dependent on the culture conditions. Finally, espH deletion showed a partial secretion defect; whereas several ESX-1 substrates were secreted in normal amounts, secretion of EsxA and EsxB was diminished and secretion of EspE and EspF was fully blocked. Interaction studies showed that EspH binds EspE and therefore could function as a specific chaperone for this substrate. Despite the observed differences in secretion, hemolytic activity was lost in all M. marinum mutants, implying that hemolytic activity is not strictly correlated with EsxA secretion. Surprisingly, while EspH is essential for successful infection of phagocytic host cells, deletion of espH resulted in a significantly increased virulence phenotype in zebrafish larvae, linked to poor granuloma formation and extracellular outgrowth. Together, these data show that different sets of ESX-1 substrates play different roles at various steps of the infection cycle of M. marinum.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium marinum/metabolismo , Mycobacterium marinum/patogenicidad , Sistemas de Secreción Tipo VII/genética , Factores de Virulencia/fisiología , Animales , Proteínas Bacterianas/genética , Células Cultivadas , Embrión no Mamífero , Larva , Ratones , Mycobacterium marinum/genética , Células RAW 264.7 , Ovinos , Sistemas de Secreción Tipo VII/metabolismo , Virulencia/genética , Factores de Virulencia/genética , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo
7.
J Am Soc Mass Spectrom ; 29(8): 1571-1581, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29949055

RESUMEN

A unique method for identification of biomolecular components in different biological specimens, while preserving the capability for high speed 2D and 3D molecular imaging, is employed to investigate cellular response to oxidative stress. The employed method enables observing the distribution of the antioxidant α-tocopherol and other molecules in cellular structures via time-of-flight secondary ion mass spectrometry (TOF-SIMS (MS1)) imaging in parallel with tandem mass spectrometry (MS2) imaging, collected simultaneously. The described method is employed to examine a network formed by neuronal cells differentiated from human induced pluripotent stem cells (iPSCs), a model for investigating human neurons in vitro. The antioxidant α-tocopherol is identified in situ within different cellular layers utilizing a 3D TOF-SIMS tandem MS imaging analysis. As oxidative stress also plays an important role in mediating inflammation, the study was expanded to whole body tissue sections of M. marinum-infected zebrafish, a model organism for tuberculosis. The TOF-SIMS tandem MS imaging results reveal an increased presence of α-tocopherol in response to the pathogen. Graphical Abstract ᅟ.


Asunto(s)
Imagen Molecular/métodos , Espectrometría de Masas en Tándem/métodos , alfa-Tocoferol/análisis , Animales , Células Cultivadas , Peces , Humanos , Imagenología Tridimensional , Células Madre Pluripotentes Inducidas/química , Masculino , Persona de Mediana Edad , Neuronas/química , Imagen de Cuerpo Entero/métodos , Pez Cebra
8.
Cell Microbiol ; 20(9): e12858, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29749044

RESUMEN

Central nervous system (CNS) infection by Mycobacterium tuberculosis is one of the most devastating complications of tuberculosis, in particular in early childhood. In order to induce CNS infection, M. tuberculosis needs to cross specialised barriers protecting the brain. How M. tuberculosis crosses the blood-brain barrier (BBB) and enters the CNS is not well understood. Here, we use transparent zebrafish larvae and the closely related pathogen Mycobacterium marinum to answer this question. We show that in the early stages of development, mycobacteria rapidly infect brain tissue, either as free mycobacteria or within circulating macrophages. After the formation of a functionally intact BBB, the infiltration of brain tissue by infected macrophages is delayed, but not blocked, suggesting that crossing the BBB via phagocytic cells is one of the mechanisms used by mycobacteria to invade the CNS. Interestingly, depletion of phagocytic cells did not prevent M. marinum from infecting the brain tissue, indicating that free mycobacteria can independently cause brain infection. Detailed analysis showed that mycobacteria are able to cause vasculitis by extracellular outgrowth in the smaller blood vessels and by infecting endothelial cells. Importantly, we could show that this second mechanism is an active process that depends on an intact ESX-1 secretion system, which extends the role of ESX-1 secretion beyond the macrophage infection cycle.


Asunto(s)
Barrera Hematoencefálica/microbiología , Infecciones del Sistema Nervioso Central/patología , Interacciones Huésped-Patógeno , Infecciones por Mycobacterium no Tuberculosas/patología , Mycobacterium marinum/crecimiento & desarrollo , Animales , Encéfalo/microbiología , Modelos Animales de Enfermedad , Macrófagos/microbiología , Pez Cebra
10.
J Neuroinflammation ; 13(1): 188, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27542968

RESUMEN

BACKGROUND: Streptococcus pneumoniae is one of the most important causes of bacterial meningitis, an infection where unfavourable outcome is driven by bacterial and host-derived toxins. In this study, we developed and characterized a pneumococcal meningitis model in zebrafish embryos that allows for real-time investigation of early host-microbe interaction. METHODS: Zebrafish embryos were infected in the caudal vein or hindbrain ventricle with green fluorescent wild-type S. pneumoniae D39 or a pneumolysin-deficient mutant. The kdrl:mCherry transgenic zebrafish line was used to visualize the blood vessels, whereas phagocytic cells were visualized by staining with far red anti-L-plastin or in mpx:GFP/mpeg1:mCherry zebrafish, that have green fluorescent neutrophils and red fluorescent macrophages. Imaging was performed by fluorescence confocal and time-lapse microscopy. RESULTS: After infection by caudal vein, we saw focal clogging of the pneumococci in the blood vessels and migration of bacteria through the blood-brain barrier into the subarachnoid space and brain tissue. Infection with pneumolysin-deficient S. pneumoniae in the hindbrain ventricle showed attenuated growth and migration through the brain as compared to the wild-type strain. Time-lapse and confocal imaging revealed that the initial innate immune response to S. pneumoniae in the subarachnoid space mainly consisted of neutrophils and that pneumolysin-mediated cytolytic activity caused a marked reduction of phagocytes. CONCLUSIONS: This new meningitis model permits detailed analysis and visualization of host-microbe interaction in pneumococcal meningitis in real time and is a very promising tool to further our insights in the pathogenesis of pneumococcal meningitis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Inmunidad Innata/fisiología , Meningitis Neumocócica/microbiología , Meningitis Neumocócica/patología , Streptococcus pneumoniae/patogenicidad , Factores de Edad , Animales , Animales Modificados Genéticamente , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Embrión no Mamífero/microbiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Meningitis Neumocócica/genética , Meningitis Neumocócica/mortalidad , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteína Fluorescente Roja
11.
Dev Comp Immunol ; 60: 108-14, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26920462

RESUMEN

Chicken cathelicidin-2 (CATH-2) is a host defense peptide that exhibits immunomodulatory and antibacterial properties. Here we examined effects of CATH-2 in zebrafish embryos in the absence and presence of infection. Yolk-injection of 0.2-1.5 h post-fertilized (hpf) zebrafish embryos with 2.6 ng/kg CATH-2 increased proliferation of phagocytic cells at 48 hpf by 30%. A lethal infection model was developed to test the prophylactic protective effect of CATH-2 peptide. Embryos (0.2-1.5 hpf) were injected with 2.6 ng/kg CATH-2, challenged with a lethal dose of fluorescently labeled Salmonella enteritidis pGMDs3 at 28 hpf and monitored for survival. Prophylactic treatment with CATH-2 was found to delay infection starting at 22 h post-infection (hpi). At 18-20 hpi, significantly lower (2-fold) fluorescence intensity and decreased bacterial loads were detected in peptide-treated embryos. Thus prophylactic administration of low CATH-2 concentrations confer partial protection in zebrafish embryos by boosting the innate immune system.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Enfermedades de los Peces/inmunología , Inmunidad Innata/efectos de los fármacos , Salmonelosis Animal/inmunología , Pez Cebra/inmunología , Animales , Proliferación Celular , Evaluación Preclínica de Medicamentos , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/inmunología , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/prevención & control , Fagocitos/fisiología , Salmonelosis Animal/microbiología , Salmonelosis Animal/prevención & control , Salmonella enteritidis/inmunología
12.
PLoS One ; 10(6): e0126378, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26083432

RESUMEN

Adaptive immunity in homeotherms depends greatly on CD4+ Th cells which release cytokines in response to specific antigen stimulation. Whilst bony fish and poikilothermic tetrapods possess cells that express TcR and CD4-related genes (that exist in two forms in teleost fish; termed CD4-1 and CD4-2), to date there is no unequivocal demonstration that cells equivalent to Th exist. Thus, in this study we determined whether CD4-1+ lymphocytes can express cytokines typical of Th cells following antigen specific stimulation, using the zebrafish (Danio rerio). Initially, we analyzed the CD4 locus in zebrafish and found three CD4 homologues, a CD4-1 molecule and two CD4-2 molecules. The zfCD4-1 and zfCD4-2 transcripts were detected in immune organs and were most highly expressed in lymphocytes. A polyclonal antibody to zfCD4-1 was developed and used with an antibody to ZAP70 and revealed double positive cells by immunohistochemistry, and in the Mycobacterium marinum disease model CD4-1+ cells were apparent surrounding the granulomas typical of the infection. Next a prime-boost experiment, using human gamma globulin as antigen, was performed and revealed for the first time in fish that zfCD4-1+ lymphocytes increase the expression of cytokines and master transcription factors relevant to Th1/Th2-type responses as a consequence of boosting with specific antigen.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Antígenos de Histocompatibilidad Clase II/inmunología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Infecciones por Mycobacterium no Tuberculosas/veterinaria , ARN Mensajero/inmunología , Inmunidad Adaptativa , Secuencia de Aminoácidos , Animales , Anticuerpos/química , Linfocitos T CD4-Positivos/clasificación , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/patología , Citocinas/inmunología , Sitios Genéticos/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Infecciones por Mycobacterium no Tuberculosas/genética , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/inmunología , Filogenia , ARN Mensajero/genética , Alineación de Secuencia , Balance Th1 - Th2 , Proteína Tirosina Quinasa ZAP-70/genética , Proteína Tirosina Quinasa ZAP-70/inmunología , Pez Cebra/clasificación , Pez Cebra/genética , Pez Cebra/inmunología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/inmunología , gammaglobulinas/administración & dosificación
13.
Infect Immun ; 83(5): 1778-88, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25690095

RESUMEN

The interaction of environmental bacteria with unicellular eukaryotes is generally considered a major driving force for the evolution of intracellular pathogens, allowing them to survive and replicate in phagocytic cells of vertebrate hosts. To test this hypothesis on a genome-wide level, we determined for the intracellular pathogen Mycobacterium marinum whether it uses conserved strategies to exploit host cells from both protozoan and vertebrate origin. Using transposon-directed insertion site sequencing (TraDIS), we determined differences in genetic requirements for survival and replication in phagocytic cells of organisms from different kingdoms. In line with the general hypothesis, we identified a number of general virulence mechanisms, including the type VII protein secretion system ESX-1, biosynthesis of polyketide lipids, and utilization of sterols. However, we were also able to show that M. marinum contains an even larger set of host-specific virulence determinants, including proteins involved in the modification of surface glycolipids and, surprisingly, the auxiliary proteins of the ESX-1 system. Several of these factors were in fact counterproductive in other hosts. Therefore, M. marinum contains different sets of virulence factors that are tailored for specific hosts. Our data imply that although amoebae could function as a training ground for intracellular pathogens, they do not fully prepare pathogens for crossing species barriers.


Asunto(s)
Genoma Bacteriano , Viabilidad Microbiana , Mutagénesis Insercional , Mycobacterium marinum/genética , Mycobacterium marinum/fisiología , Factores de Virulencia/metabolismo , Acanthamoeba castellanii/microbiología , Animales , Elementos Transponibles de ADN , Dictyostelium/microbiología , Humanos , Mycobacterium marinum/crecimiento & desarrollo , Fagocitos/microbiología , Virulencia , Factores de Virulencia/genética
14.
Dis Model Mech ; 8(3): 253-69, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25573892

RESUMEN

The recruitment of leukocytes to infectious foci depends strongly on the local release of chemoattractant mediators. The human CXC chemokine receptor 3 (CXCR3) is an important node in the chemokine signaling network and is expressed by multiple leukocyte lineages, including T cells and macrophages. The ligands of this receptor originate from an ancestral CXCL11 gene in early vertebrates. Here, we used the optically accessible zebrafish embryo model to explore the function of the CXCR3-CXCL11 axis in macrophage recruitment and show that disruption of this axis increases the resistance to mycobacterial infection. In a mutant of the zebrafish ortholog of CXCR3 (cxcr3.2), macrophage chemotaxis to bacterial infections was attenuated, although migration to infection-independent stimuli was unaffected. Additionally, attenuation of macrophage recruitment to infection could be mimicked by treatment with NBI74330, a high-affinity antagonist of CXCR3. We identified two infection-inducible CXCL11-like chemokines as the functional ligands of Cxcr3.2, showing that the recombinant proteins exerted a Cxcr3.2-dependent chemoattraction when locally administrated in vivo. During infection of zebrafish embryos with Mycobacterium marinum, a well-established model for tuberculosis, we found that Cxcr3.2 deficiency limited the macrophage-mediated dissemination of mycobacteria. Furthermore, the loss of Cxcr3.2 function attenuated the formation of granulomatous lesions, the typical histopathological features of tuberculosis, and led to a reduction in the total bacterial burden. Prevention of mycobacterial dissemination by targeting the CXCR3 pathway, therefore, might represent a host-directed therapeutic strategy for treatment of tuberculosis. The demonstration of a conserved CXCR3-CXCL11 signaling axis in zebrafish extends the translational applicability of this model for studying diseases involving the innate immune system.


Asunto(s)
Quimiocina CXCL11/metabolismo , Macrófagos/metabolismo , Infecciones por Mycobacterium/metabolismo , Receptores CXCR3/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Animales , Movimiento Celular , Factores Quimiotácticos/farmacología , Codón sin Sentido/genética , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Granuloma/patología , Humanos , Larva/crecimiento & desarrollo , Macrófagos/efectos de los fármacos , Infecciones por Mycobacterium/microbiología , Infecciones por Mycobacterium/patología , Fagocitos/metabolismo , Receptores CXCR3/análisis , Receptores CXCR3/antagonistas & inhibidores , Receptores CXCR3/genética , Proteínas Recombinantes/farmacología , Pez Cebra/embriología , Pez Cebra/microbiología , Proteínas de Pez Cebra/análisis , Proteínas de Pez Cebra/genética
15.
Cold Spring Harb Perspect Med ; 5(3): a018580, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25414379

RESUMEN

Over the past decade the zebrafish (Danio rerio) has become an attractive new vertebrate model organism for studying mycobacterial pathogenesis. The combination of medium-throughput screening and real-time in vivo visualization has allowed new ways to dissect host pathogenic interaction in a vertebrate host. Furthermore, genetic screens on the host and bacterial sides have elucidated new mechanisms involved in the initiation of granuloma formation and the importance of a balanced immune response for control of mycobacterial pathogens. This article will highlight the unique features of the zebrafish-Mycobacterium marinum infection model and its added value for tuberculosis research.


Asunto(s)
Modelos Animales de Enfermedad , Tuberculosis/fisiopatología , Pez Cebra/microbiología , Animales , Infecciones por Mycobacterium no Tuberculosas/fisiopatología
16.
Dis Model Mech ; 7(9): 1111-22, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24997190

RESUMEN

Tuberculous meningitis (TBM) is one of the most severe extrapulmonary manifestations of tuberculosis, with a high morbidity and mortality. Characteristic pathological features of TBM are Rich foci, i.e. brain- and spinal-cord-specific granulomas formed after hematogenous spread of pulmonary tuberculosis. Little is known about the early pathogenesis of TBM and the role of Rich foci. We have adapted the zebrafish model of Mycobacterium marinum infection (zebrafish-M. marinum model) to study TBM. First, we analyzed whether TBM occurs in adult zebrafish and showed that intraperitoneal infection resulted in granuloma formation in the meninges in 20% of the cases, with occasional brain parenchyma involvement. In zebrafish embryos, bacterial infiltration and clustering of infected phagocytes was observed after infection at three different inoculation sites: parenchyma, hindbrain ventricle and caudal vein. Infection via the bloodstream resulted in the formation of early granulomas in brain tissue in 70% of the cases. In these zebrafish embryos, infiltrates were located in the proximity of blood vessels. Interestingly, no differences were observed when embryos were infected before or after early formation of the blood-brain barrier (BBB), indicating that bacteria are able to cross this barrier with relatively high efficiency. In agreement with this observation, infected zebrafish larvae also showed infiltration of the brain tissue. Upon infection of embryos with an M. marinum ESX-1 mutant, only small clusters and scattered isolated phagocytes with high bacterial loads were present in the brain tissue. In conclusion, our adapted zebrafish-M. marinum infection model for studying granuloma formation in the brain will allow for the detailed analysis of both bacterial and host factors involved in TBM. It will help solve longstanding questions on the role of Rich foci and potentially contribute to the development of better diagnostic tools and therapeutics.


Asunto(s)
Modelos Animales de Enfermedad , Mycobacterium marinum/fisiología , Tuberculosis Meníngea/microbiología , Animales , Barrera Hematoencefálica , Infecciones por Mycobacterium no Tuberculosas/microbiología , Pez Cebra
17.
Methods Mol Biol ; 1149: 709-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24818945

RESUMEN

In the last years, the zebrafish (Danio rerio) has become an important vertebrate animal model to study host-pathogen interactions, especially in its embryonic stage. The presence of a fully developed innate immune system in the first days of embryogenesis, the facility of obtaining and manipulating large numbers of embryos, the optical transparency of the embryos that allow the direct visualization of bacterial infections, a wide range of genetic tools, and extensive mutant resources and collections of transgenic reporter lines are important advantages of the zebrafish-embryo model. Pseudomonas aeruginosa is able to lethally infect zebrafish embryos when the amount of cells injected exceeds the phagocytic capacity of the embryo. Different studies have proved the suitability of zebrafish embryos as a model to analyze P. aeruginosa infection. Here we describe the detailed protocols to establish a P. aeruginosa infection in zebrafish embryos and to image the interaction of the bacterium with this host with fluorescent microscopy.


Asunto(s)
Bioensayo/métodos , Interacciones Huésped-Patógeno , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Pez Cebra/microbiología , Animales , Embrión no Mamífero/microbiología , Imagenología Tridimensional , Inyecciones , Mamíferos , Virulencia , Pez Cebra/embriología
18.
Structure ; 22(5): 719-30, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24704253

RESUMEN

The cell-envelope of Mycobacterium tuberculosis plays a key role in bacterial virulence and antibiotic resistance. Little is known about the molecular mechanisms of regulation of cell-envelope formation. Here, we elucidate functional and structural properties of RNase AS, which modulates M. tuberculosis cell-envelope properties and strongly impacts bacterial virulence in vivo. The structure of RNase AS reveals a resemblance to RNase T from Escherichia coli, an RNase of the DEDD family involved in RNA maturation. We show that RNase AS acts as a 3'-5'-exoribonuclease that specifically hydrolyzes adenylate-containing RNA sequences. Also, crystal structures of complexes with AMP and UMP reveal the structural basis for the observed enzyme specificity. Notably, RNase AS shows a mechanism of substrate recruitment, based on the recognition of the hydrogen bond donor NH2 group of adenine. Our work opens a field for the design of drugs able to reduce bacterial virulence in vivo.


Asunto(s)
Mycobacterium tuberculosis/patogenicidad , Ribonucleasas/química , Ribonucleasas/metabolismo , Adenina , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Embrión no Mamífero/microbiología , Exorribonucleasas/química , Técnicas de Inactivación de Genes , Enlace de Hidrógeno , Modelos Moleculares , Mutación , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/enzimología , Poli A/metabolismo , Multimerización de Proteína , Ribonucleasas/genética , Especificidad por Sustrato , Uridina Monofosfato/química , Uridina Monofosfato/metabolismo , Pez Cebra/embriología , Pez Cebra/microbiología
19.
EMBO Mol Med ; 6(3): 372-83, 2014 03.
Artículo en Inglés | MEDLINE | ID: mdl-24500695

RESUMEN

The benzothiazinone lead compound, BTZ043, kills Mycobacterium tuberculosis by inhibiting the essential flavo-enzyme DprE1, decaprenylphosphoryl-beta-D-ribose 2-epimerase. Here, we synthesized a new series of piperazine-containing benzothiazinones (PBTZ) and show that, like BTZ043, the preclinical candidate PBTZ169 binds covalently to DprE1. The crystal structure of the DprE1-PBTZ169 complex reveals formation of a semimercaptal adduct with Cys387 in the active site and explains the irreversible inactivation of the enzyme. Compared to BTZ043, PBTZ169 has improved potency, safety and efficacy in zebrafish and mouse models of tuberculosis (TB). When combined with other TB drugs, PBTZ169 showed additive activity against M. tuberculosis in vitro except with bedaquiline (BDQ) where synergy was observed. A new regimen comprising PBTZ169, BDQ and pyrazinamide was found to be more efficacious than the standard three drug treatment in a murine model of chronic disease. PBTZ169 is thus an attractive drug candidate to treat TB in humans.


Asunto(s)
Antituberculosos/uso terapéutico , Compuestos de Espiro/uso terapéutico , Tiazinas/uso terapéutico , Tuberculosis/tratamiento farmacológico , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/metabolismo , Animales , Antituberculosos/síntesis química , Antituberculosos/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Embrión no Mamífero/efectos de los fármacos , Células Hep G2 , Humanos , Pulmón/metabolismo , Ratones , Simulación de Dinámica Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Piperazinas/química , Piperazinas/farmacología , Piperazinas/uso terapéutico , Compuestos de Espiro/química , Compuestos de Espiro/farmacocinética , Compuestos de Espiro/farmacología , Bazo/metabolismo , Tiazinas/química , Tiazinas/farmacocinética , Tiazinas/farmacología , Pez Cebra/crecimiento & desarrollo
20.
Cell Microbiol ; 16(2): 280-95, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24119166

RESUMEN

The pathogenicity of mycobacteria is closely associated with their ability to export virulence factors. For this purpose, mycobacteria possess different protein secretion systems, including the accessory Sec translocation pathway, SecA2. Although this pathway is associated with intracellular survival and virulence, the SecA2-dependent effector proteins remain largely undefined. In this work, we studied a Mycobacterium marinum secA2 mutant with an impaired capacity to initiate granuloma formation in zebrafish embryos. By comparing the proteomic profile of cell envelope fractions from the secA2 mutant with wild type M. marinum, we identified putative SecA2-dependent substrates. Immunoblotting procedures confirmed SecA2-dependent membrane localization for several of these proteins, including the virulence factor protein kinase G (PknG). Interestingly, phenotypical defects of the secA2 mutant are similar to those described for ΔpknG, including phagosomal maturation. Overexpression of PknG in the secA2 mutant restored its localization to the cell envelope. Importantly, PknG-overexpression also partially restored the virulence of the secA2 mutant, as indicated by enhanced infectivity in zebrafish embryos and restored inhibition of phagosomal maturation. These results suggest that SecA2-dependent membrane localization of PknG is an important determinant for M. marinum virulence.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Mycobacterium marinum/metabolismo , Factores de Virulencia/metabolismo , Animales , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Immunoblotting , Mutagénesis Insercional , Infecciones por Mycobacterium/microbiología , Mycobacterium marinum/patogenicidad , Especificidad por Sustrato , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA