Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Microbiol ; 117(3): 682-692, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34605588

RESUMEN

Respiratory infections remain a major global health concern. Tuberculosis is one of the top 10 causes of death worldwide, while infections with Non-Tuberculous Mycobacteria are rising globally. Recent advances in human tissue modeling offer a unique opportunity to grow different human "organs" in vitro, including the human airway, that faithfully recapitulates lung architecture and function. Here, we have explored the potential of human airway organoids (AOs) as a novel system in which to assess the very early steps of mycobacterial infection. We reveal that Mycobacterium tuberculosis (Mtb) and Mycobacterium abscessus (Mabs) mainly reside as extracellular bacteria and infect epithelial cells with very low efficiency. While the AO microenvironment was able to control, but not eliminate Mtb, Mabs thrives. We demonstrate that AOs responded to infection by modulating cytokine, antimicrobial peptide, and mucin gene expression. Given the importance of myeloid cells in mycobacterial infection, we co-cultured infected AOs with human monocyte-derived macrophages and found that these cells interact with the organoid epithelium. We conclude that adult stem cell (ASC)-derived AOs can be used to decipher very early events of mycobacteria infection in human settings thus offering new avenues for fundamental and therapeutic research.


Asunto(s)
Mycobacterium abscessus , Mycobacterium tuberculosis , Tuberculosis , Humanos , Macrófagos/microbiología , Micobacterias no Tuberculosas , Organoides , Tuberculosis/microbiología
2.
Cell Microbiol ; 23(7): e13344, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33860624

RESUMEN

The interactions between microbes and their hosts are among the most complex biological phenomena known today. The interaction may reach from overall beneficial interaction, as observed for most microbiome/microbiota related interactions to interaction with virulent pathogens, against which host cells have evolved sophisticated defence strategies. Among the latter, the confinement of invading pathogens in a phagosome plays a key role, which often results in the destruction of the invader, whereas some pathogens may counteract phagosomal arrest and survive by gaining access to the cytosol of the host cell. In the current review, we will discuss recent insights into this dynamic process of host-pathogen interaction, using Mycobacterium tuberculosis and related pathogenic mycobacteria as main examples.


Asunto(s)
Interacciones Huésped-Patógeno , Mycobacterium tuberculosis/patogenicidad , Fagosomas/microbiología , Tuberculosis/microbiología , Humanos , Fagocitosis
3.
Cell Microbiol ; 19(7)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28095608

RESUMEN

Although phthiocerol dimycocerosates (DIM) are major virulence factors of Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis, little is known about their mechanism of action. Localized in the outer membrane of mycobacterial pathogens, DIM are predicted to interact with host cell membranes. Interaction with eukaryotic membranes is a property shared with another virulence factor of Mtb, the early secretory antigenic target EsxA (also known as ESAT-6). This small protein, which is secreted by the type VII secretion system ESX-1 (T7SS/ESX-1), is involved in phagosomal rupture and cell death induced by virulent mycobacteria inside host phagocytes. In this work, by the use of several knock-out or knock-in mutants of Mtb or Mycobacterium bovis BCG strains and different cell biological assays, we present conclusive evidence that ESX-1 and DIM act in concert to induce phagosomal membrane damage and rupture in infected macrophages, ultimately leading to host cell apoptosis. These results identify an as yet unknown function for DIM in the infection process and open up a new research field for the study of the interaction of lipid and protein virulence factors of Mtb.


Asunto(s)
Antígenos Bacterianos/metabolismo , Apoptosis/fisiología , Proteínas Bacterianas/metabolismo , Lípidos/fisiología , Macrófagos/metabolismo , Mycobacterium bovis/patogenicidad , Mycobacterium tuberculosis/patogenicidad , Fagosomas/metabolismo , Línea Celular Tumoral , Membrana Celular/patología , Humanos , Macrófagos/microbiología , Fagosomas/microbiología , Células THP-1 , Factores de Virulencia
4.
PLoS Pathog ; 11(2): e1004650, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658322

RESUMEN

Mycobacterium tuberculosis (Mtb) uses efficient strategies to evade the eradication by professional phagocytes, involving--as recently confirmed--escape from phagosomal confinement. While Mtb determinants, such as the ESX-1 type VII secretion system, that contribute to this phenomenon are known, the host cell factors governing this important biological process are yet unexplored. Using a newly developed flow-cytometric approach for Mtb, we show that macrophages expressing the phagosomal bivalent cation transporter Nramp-1, are much less susceptible to phagosomal rupture. Together with results from the use of the phagosome acidification inhibitor bafilomycin, we demonstrate that restriction of phagosomal acidification is a prerequisite for mycobacterial phagosomal rupture and cytosolic contact. Using different in vivo approaches including an enrichment and screen for tracking rare infected phagocytes carrying the CD45.1 hematopoietic allelic marker, we here provide first and unique evidence of M. tuberculosis-mediated phagosomal rupture in mouse spleen and lungs and in numerous phagocyte types. Our results, linking the ability of restriction of phagosome acidification to cytosolic access, provide an important conceptual advance for our knowledge on host processes targeted by Mtb evasion strategies.


Asunto(s)
Sistemas de Secreción Bacterianos/inmunología , Proteínas de Transporte de Catión/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Fagosomas/inmunología , Tuberculosis/inmunología , Animales , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Macrófagos/microbiología , Ratones , Ratones Mutantes , Fagosomas/genética , Tuberculosis/genética , Tuberculosis/patología
5.
Cell Microbiol ; 18(8): 1070-7, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27247079

RESUMEN

Mycobacterium tuberculosis, the infectious agent of human tuberculosis is a master player in circumventing the defense mechanisms of the host immune system. The host-pathogen interaction in the case of an infection with M. tuberculosis is highly complex, involving dedicated mycobacterial virulence factors as well as the action of the innate and adapted immune systems, which determine the outcome of infection. Macrophages play a key role in this process through internalizing the bacterium in a phagosomal vacuole. While this action has normally the function of eliminating invading bacteria, M. tuberculosis employs efficient strategies to prevent its extermination. The question on how-and-where the bacterium succeeds in doing so has interested generations of scientists and still remains a fascinating and important research subject focused on mycobacterial lipids, secretion systems and other contributing factors. This topic is also central to the longstanding and partially controversial discussion on mycobacterial phagosomal rupture and vacuole-to-cytosol translocation, to be reviewed here in more detail.


Asunto(s)
Citosol/microbiología , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología , Vacuolas/microbiología , Animales , Interacciones Huésped-Patógeno , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Fagosomas/microbiología , Tuberculosis/inmunología
6.
PLoS Pathog ; 8(2): e1002507, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22319448

RESUMEN

Survival within macrophages is a central feature of Mycobacterium tuberculosis pathogenesis. Despite significant advances in identifying new immunological parameters associated with mycobacterial disease, some basic questions on the intracellular fate of the causative agent of human tuberculosis in antigen-presenting cells are still under debate. To get novel insights into this matter, we used a single-cell fluorescence resonance energy transfer (FRET)-based method to investigate the potential cytosolic access of M. tuberculosis and the resulting cellular consequences in an unbiased, quantitative way. Analysis of thousands of THP-1 macrophages infected with selected wild-type or mutant strains of the M. tuberculosis complex unambiguously showed that M. tuberculosis induced a change in the FRET signal after 3 to 4 days of infection, indicating phagolysosomal rupture and cytosolic access. These effects were not seen for the strains M. tuberculosisΔRD1 or BCG, both lacking the ESX-1 secreted protein ESAT-6, which reportedly shows membrane-lysing properties. Complementation of these strains with the ESX-1 secretion system of M. tuberculosis restored the ability to cause phagolysosomal rupture. In addition, control experiments with the fish pathogen Mycobacterium marinum showed phagolysosomal translocation only for ESX-1 intact strains, further validating our experimental approach. Most importantly, for M. tuberculosis as well as for M. marinum we observed that phagolysosomal rupture was followed by necrotic cell death of the infected macrophages, whereas ESX-1 deletion- or truncation-mutants that remained enclosed within phagolysosomal compartments did not induce such cytotoxicity. Hence, we provide a novel mechanism how ESX-1 competent, virulent M. tuberculosis and M. marinum strains induce host cell death and thereby escape innate host defenses and favor their spread to new cells. In this respect, our results also open new research directions in relation with the extracellular localization of M. tuberculosis inside necrotic lesions that can now be tackled from a completely new perspective.


Asunto(s)
Macrófagos/metabolismo , Infecciones por Mycobacterium/patología , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Fagosomas/patología , Muerte Celular , Línea Celular , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas de Homeodominio/metabolismo , Humanos , Evasión Inmune , Macrófagos/microbiología , Infecciones por Mycobacterium/inmunología , Infecciones por Mycobacterium/metabolismo , Mycobacterium marinum/inmunología , Mycobacterium marinum/metabolismo , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/metabolismo , Fagosomas/metabolismo , Fagosomas/microbiología , Salmonella typhimurium/patogenicidad , Shigella flexneri/patogenicidad
7.
Mol Microbiol ; 83(6): 1195-209, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22340629

RESUMEN

The chromosome of Mycobacterium tuberculosis encodes five type VII secretion systems (ESX-1-ESX-5). While the role of the ESX-1 and ESX-3 systems in M. tuberculosis has been elucidated, predictions for the function of the ESX-5 system came from data obtained in Mycobacterium marinum, where it transports PPE and PE_PGRS proteins and modulates innate immune responses. To define the role of the ESX-5 system in M. tuberculosis, in this study, we have constructed five M. tuberculosis H37Rv ESX-5 knockout/deletion mutants, inactivating eccA(5), eccD(5), rv1794 and esxM genes or the ppe25-pe19 region. Whereas the Mtbrv1794ko displayed no obvious phenotype, the other four mutants showed defects in secretion of the ESX-5-encoded EsxN and PPE41, a representative member of the large PPE protein family. Strikingly, the MtbeccD(5) ko mutant also showed enhanced sensitivity to detergents and hydrophilic antibiotics. When the virulence of the five mutants was evaluated, the MtbeccD(5) ko and MtbΔppe25-pe19 mutants were found attenuated both in macrophages and in the severe combined immune-deficient mouse infection model. Altogether these findings indicate an essential role of ESX-5 for transport of PPE proteins, cell wall integrity and full virulence of M. tuberculosis, thereby opening interesting new perspectives for the study of this human pathogen.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Pared Celular/metabolismo , Mycobacterium tuberculosis/metabolismo , Tuberculosis/microbiología , Secuencias de Aminoácidos , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Pared Celular/química , Pared Celular/genética , Células Cultivadas , Humanos , Macrófagos/microbiología , Ratones , Ratones SCID , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Transporte de Proteínas , Virulencia
8.
J Biol Chem ; 286(30): 26987-95, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21646350

RESUMEN

Autophagy is an important mechanism of innate immune defense. We have recently shown that autophagy components are recruited with septins, a new and increasingly characterized cytoskeleton component, to intracytosolic Shigella that have started to polymerize actin. On the other hand, intracytosolic Listeria avoids autophagy recognition by expressing ActA, a bacterial effector required for actin polymerization. Here, we exploit Shigella and Listeria as intracytosolic tools to characterize different pathways of selective autophagy. We show that the ubiquitin-binding adaptor proteins p62 and NDP52 target Shigella to an autophagy pathway dependent upon septin and actin. In contrast, p62 or NDP52 targets the Listeria ActA mutant to an autophagy pathway independent of septin or actin. TNF-α, a host cytokine produced upon bacterial infection, stimulates p62-mediated autophagic activity and restricts the survival of Shigella and the Listeria ActA mutant. These data provide a new molecular framework to understand the emerging complexity of autophagy and its ability to achieve specific clearance of intracytosolic bacteria.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Disentería Bacilar/metabolismo , Listeria monocytogenes/metabolismo , Listeriosis/metabolismo , Proteínas Nucleares/metabolismo , Shigella flexneri/metabolismo , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Bacterianas , Citosol/metabolismo , Citosol/microbiología , Disentería Bacilar/genética , Células HeLa , Humanos , Listeria monocytogenes/genética , Listeriosis/genética , Proteínas de la Membrana , Mutación , Proteínas Nucleares/genética , Septinas/genética , Septinas/metabolismo , Proteína Sequestosoma-1 , Shigella flexneri/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
Emerg Infect Dis ; 18(4): 653-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22469053

RESUMEN

The oryx bacilli are Mycobacterium tuberculosis complex organisms for which phylogenetic position and host range are unsettled. We characterized 22 isolates by molecular methods and propose elevation to subspecies status as M. orygis. M. orygis is a causative agent of tuberculosis in animals and humans from Africa and South Asia.


Asunto(s)
Genes Bacterianos , Mycobacterium tuberculosis/clasificación , Tuberculosis/microbiología , Adolescente , Adulto , Anciano , Animales , Técnicas de Tipificación Bacteriana , Niño , Preescolar , Humanos , Lactante , Persona de Mediana Edad , Tipificación de Secuencias Multilocus , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/aislamiento & purificación , Filogenia , Polimorfismo de Longitud del Fragmento de Restricción , Polimorfismo de Nucleótido Simple , Eliminación de Secuencia , Adulto Joven
10.
J Infect Dis ; 203(8): 1155-64, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21196469

RESUMEN

BACKGROUND: The pathogenesis of Mycobacterium tuberculosis largely depends on the secretion of the 6-kD early secreted antigenic target ESAT-6 (EsxA) and the 10-kD culture filtrate protein CFP-10 (EsxB) via the ESX-1/typeVII secretion system. Although gene products from the core RD1 region have been shown to be deeply implicated in this process, less is known about proteins encoded further upstream in the 5' region of the ESX-1 cluster, such as the ESX-1 secretion-associated proteins (Esps) EspF or EspG(1). METHODS: To elucidate the role of EspF/G(1), whose orthologs in Mycobacterium marinum and Mycobacterium smegmatis are reportedly involved in EsxA/B secretion, we constructed 3 M. tuberculosis knockout strains deleted for espF, espG(1) or the segment corresponding to the combined RD1(bcg)-RD1(mic) region of bacille Calmette-Guérin (BCG) and Mycobacterium microti, which also contains espF and espG(1). RESULTS: Analysis of these strains revealed that, unlike observations with the model organisms M. smegmatis or M. marinum, disruption of espF and espG(1) in M. tuberculosis did not impact the secretion and T cell recognition of EsxA/B but still caused severe attenuation. CONCLUSIONS: The separation of the 2 ESX-1-connected phenotypes (ie, EsxA/B secretion and virulence) indicates that EsxA/B secretion is not the only readout for a functional ESX-1 system and suggests that other processes involving EspF/G(1) also play important roles in ESX-1-mediated pathogenicity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Eliminación de Gen , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Ratones , Ratones Endogámicos C57BL , Familia de Multigenes , Mycobacterium tuberculosis/genética , Procesamiento Proteico-Postraduccional , Virulencia
11.
Infect Immun ; 79(7): 2829-38, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21576344

RESUMEN

Onset of the adaptive immune response in mice infected with Mycobacterium tuberculosis is accompanied by slowing of bacterial replication and establishment of a chronic infection. Stabilization of bacterial numbers during the chronic phase of infection is dependent on the activity of the gamma interferon (IFN-γ)-inducible nitric oxide synthase (NOS2). Previously, we described a differential signature-tagged mutagenesis screen designed to identify M. tuberculosis "counterimmune" mechanisms and reported the isolation of three mutants in the H37Rv strain background containing transposon insertions in the rv0072, rv0405, and rv2958c genes. These mutants were impaired for replication and virulence in NOS2(-/-) mice but were growth-proficient and virulent in IFN-γ(-/-) mice, suggesting that the disrupted genes were required for bacterial resistance to an IFN-γ-dependent immune mechanism other than NOS2. Here, we report that the attenuation of these strains is attributable to an underlying transposon-independent deficiency in biosynthesis of phthiocerol dimycocerosate (PDIM), a cell wall lipid that is required for full virulence in mice. We performed whole-genome resequencing of a PDIM-deficient clone and identified a spontaneous point mutation in the putative polyketide synthase PpsD that results in a G44C amino acid substitution. We demonstrate by complementation with the wild-type ppsD gene and reversion of the ppsD gene to the wild-type sequence that the ppsD(G44C) point mutation is responsible for PDIM deficiency, virulence attenuation in NOS2(-/-) and wild-type C57BL/6 mice, and a growth advantage in vitro in liquid culture. We conclude that PDIM biosynthesis is required for M. tuberculosis resistance to an IFN-γ-mediated immune response that is independent of NOS2.


Asunto(s)
Interferón gamma/inmunología , Lípidos/biosíntesis , Mycobacterium tuberculosis/inmunología , Sintasas Poliquetidas/genética , Inmunidad Adaptativa , Sustitución de Aminoácidos , Animales , Pared Celular/química , Elementos Transponibles de ADN , Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/patogenicidad , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Mutación Puntual , Sintasas Poliquetidas/química , Sintasas Poliquetidas/metabolismo
12.
Bioessays ; 31(4): 378-88, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19274661

RESUMEN

In contrast to the great majority of mycobacterial species that are harmless saprophytes, Mycobacterium tuberculosis and other closely related tubercle bacilli have evolved to be among the most important human and animal pathogens. The need to develop new strategies in the fight against tuberculosis (TB) and related diseases has fuelled research into the evolutionary success of the M. tuberculosis complex members. Amongst the various disciplines, genomics and functional genomics have been instrumental in improving our understanding of these organisms. In this review we will present some of the recent key findings on molecular determinants of mycobacterial pathogenicity and attenuation, the evolution of M. tuberculosis, genome dynamics, antigen mining for improved diagnostic and subunit antigens, and finally the identification of novel drug targets. The genomics revolution has changed the landscape of TB research, and now underpins our renewed efforts to defeat this deadly pathogen.


Asunto(s)
Evolución Biológica , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Animales , Genoma Bacteriano/genética , Humanos , Mycobacterium tuberculosis/genética , Tuberculosis/diagnóstico , Tuberculosis/tratamiento farmacológico , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Factores de Virulencia/fisiología
13.
Artículo en Inglés | MEDLINE | ID: mdl-32923411

RESUMEN

Mycobacterium tuberculosis (Mtb) synthesizes a variety of atypical lipids that are exposed at the cell surface and help the bacterium infect macrophages and escape elimination by the cell's immune responses. In the present study, we investigate the mechanism of action of one family of hydrophobic lipids, the phthiocerol dimycocerosates (DIM/PDIM), major lipid virulence factors. DIM are transferred from the envelope of Mtb to host membranes during infection. Using the polarity-sensitive fluorophore C-Laurdan, we visualized that DIM decrease the membrane polarity of a supported lipid bilayer put in contact with mycobacteria, even beyond the site of contact. We observed that DIM activate the complement receptor 3, a predominant receptor for phagocytosis of Mtb by macrophages. DIM also increased the activity of membrane-permeabilizing effectors of Mtb, among which the virulence factor EsxA. This is consistent with previous observations that DIM help Mtb disrupt host cell membranes. Taken together, our data show that transferred DIM spread within the target membrane, modify its physical properties and increase the activity of host cell receptors and bacterial effectors, diverting in a non-specific manner host cell functions. We therefore bring new insight into the molecular mechanisms by which DIM increase Mtb's capability to escape the cell's immune responses.


Asunto(s)
Mycobacterium tuberculosis , Lípidos , Macrófagos , Fagocitosis
14.
FEBS J ; 274(8): 1957-69, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17371506

RESUMEN

Phthiocerol dimycocerosates and related compounds are important molecules in the biology of Mycobacterium tuberculosis, playing a key role in the permeability barrier and in pathogenicity. Both phthiocerol dimycocerosates, the major compounds, and phthiodiolone dimycocerosates, the minor constituents, are found in the cell envelope of M. tuberculosis, but their specific roles in the biology of the tubercle bacillus have not been established yet. According to the current model of their biosynthesis, phthiocerol is produced from phthiodiolone through a two-step process in which the keto group is first reduced and then methylated. We have previously identified the methyltransferase enzyme that is involved in this process, encoded by the gene Rv2952 in M. tuberculosis. In this study, we report the construction and biochemical analyses of an M. tuberculosis strain mutated in gene Rv2951c. This mutation prevents the formation of phthiocerol and phenolphthiocerol derivatives, but leads to the accumulation of phthiodiolone dimycocerosates and glycosylated phenolphthiodiolone dimycocerosates. These results provide the formal evidence that Rv2951c encodes the ketoreductase catalyzing the reduction of phthiodiolone and phenolphthiodiolone to yield phthiotriol and phenolphthiotriol, which are the substrates of the methyltransferase encoded by gene Rv2952. We also compared the resistance to SDS and replication in mice of the Rv2951c mutant, deficient in synthesis of phthiocerol dimycocerosates but producing phthiodiolone dimycocerosates, with those of a wild-type strain and a mutant without phthiocerol and phthiodiolone dimycocerosates. The results established the functional redundancy between phthiocerol and phthiodiolone dimycocerosates in both the protection of the mycobacterial cell and the pathogenicity of M. tuberculosis in mice.


Asunto(s)
Lípidos/fisiología , Mycobacterium tuberculosis/patogenicidad , Animales , Secuencia de Bases , Lípidos/química , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/metabolismo , Permeabilidad , Dodecil Sulfato de Sodio/farmacología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Relación Estructura-Actividad , Virulencia
15.
Front Microbiol ; 8: 2284, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29218036

RESUMEN

Mycobacterium tuberculosis (Mtb), the etiological agent of human tuberculosis (TB), has plagued humans for thousands of years. TB still remains a major public health problem in our era, causing more than 4,400 deaths worldwide every day and killing more people than HIV. After inhaling Mtb-contaminated aerosols, TB primo-infection starts in the terminal lung airways, where Mtb is taken up by alveolar macrophages. Although macrophages are known as professional killers for pathogens, Mtb has adopted remarkable strategies to circumvent host defenses, building suitable conditions to survive and proliferate. Within macrophages, Mtb initially resides inside phagosomes, where its survival mostly depends on its ability to take control of phagosomal processing, through inhibition of phagolysosome biogenesis and acidification processes, and by progressively getting access to the cytosol. Bacterial access to the cytosolic space is determinant for specific immune responses and cell death programs, both required for the replication and the dissemination of Mtb. Comprehension of the molecular events governing Mtb survival within macrophages is fundamental for the improvement of vaccine-based and therapeutic strategies in order to help the host to better defend itself in the battle against the fierce invader Mtb. In this mini-review, we discuss recent research exploring how Mtb conquers and transforms the macrophage into a strategic base for its survival and dissemination as well as the associated defense strategies mounted by host.

16.
Cell Rep ; 18(11): 2752-2765, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28297677

RESUMEN

Recent insights into the mechanisms by which Mycobacterium tuberculosis, the etiologic agent of human tuberculosis, is recognized by cytosolic nucleotide sensors have opened new avenues for rational vaccine design. The only licensed anti-tuberculosis vaccine, Mycobacterium bovis BCG, provides limited protection. A feature of BCG is the partial deletion of the ESX-1 type VII secretion system, which governs phagosomal rupture and cytosolic pattern recognition, key intracellular phenotypes linked to increased immune signaling. Here, by heterologously expressing the esx-1 region of Mycobacterium marinum in BCG, we engineered a low-virulence, ESX-1-proficient, recombinant BCG (BCG::ESX-1Mmar) that induces the cGas/STING/TBK1/IRF-3/type I interferon axis and enhances AIM2 and NLRP3 inflammasome activity, resulting in both higher proportions of CD8+ T cell effectors against mycobacterial antigens shared with BCG and polyfunctional CD4+ Th1 cells specific to ESX-1 antigens. Importantly, independent mouse vaccination models show that BCG::ESX-1Mmar confers superior protection relative to parental BCG against challenges with highly virulent M. tuberculosis.


Asunto(s)
Vacuna BCG/inmunología , Proteínas Bacterianas/metabolismo , Citosol/inmunología , Mycobacterium marinum/patogenicidad , Transducción de Señal , Tuberculosis/inmunología , Tuberculosis/prevención & control , Vacunas Sintéticas/inmunología , Animales , Prueba de Complementación Genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Inmunización , Ratones SCID , Fagosomas/metabolismo , Células TH1/inmunología , Tuberculosis/microbiología , Virulencia
17.
Nat Rev Microbiol ; 14(11): 677-691, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27665717

RESUMEN

Mycobacterium tuberculosis uses sophisticated secretion systems, named 6 kDa early secretory antigenic target (ESAT6) protein family secretion (ESX) systems (also known as type VII secretion systems), to export a set of effector proteins that helps the pathogen to resist or evade the host immune response. Since the discovery of the esx loci during the M. tuberculosis H37Rv genome project, structural biology, cell biology and evolutionary analyses have advanced our knowledge of the function of these systems. In this Review, we highlight the intriguing roles that these studies have revealed for ESX systems in bacterial survival and pathogenicity during infection with M. tuberculosis. Furthermore, we discuss the diversity of ESX systems that has been described among mycobacteria and selected non-mycobacterial species. Finally, we consider how our knowledge of ESX systems might be applied to the development of novel strategies for the treatment and prevention of disease.


Asunto(s)
Proteínas Bacterianas/metabolismo , Evolución Molecular , Interacciones Huésped-Patógeno , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Sistemas de Secreción Tipo VII/fisiología , Animales , Antígenos Bacterianos , Proteínas Bacterianas/genética , Transporte Biológico , Humanos , Mycobacterium tuberculosis/fisiología , Transporte de Proteínas , Tuberculosis/inmunología , Tuberculosis/prevención & control , Tuberculosis/terapia , Sistemas de Secreción Tipo VII/genética
18.
Open Biol ; 6(11)2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27906132

RESUMEN

Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium responsible for pulmonary and cutaneous infections in immunocompetent patients and in patients with Mendelian disorders, such as cystic fibrosis (CF). Mycobacterium abscessus is known to transition from a smooth (S) morphotype with cell surface-associated glycopeptidolipids (GPL) to a rough (R) morphotype lacking GPL. Herein, we show that M. abscessus S and R variants are able to grow inside macrophages and are present in morphologically distinct phagosomes. The S forms are found mostly as single bacteria within phagosomes characterized by a tightly apposed phagosomal membrane and the presence of an electron translucent zone (ETZ) surrounding the bacilli. By contrast, infection with the R form leads to phagosomes often containing more than two bacilli, surrounded by a loose phagosomal membrane and lacking the ETZ. In contrast to the R variant, the S variant is capable of restricting intraphagosomal acidification and induces less apoptosis and autophagy. Importantly, the membrane of phagosomes enclosing the S forms showed signs of alteration, such as breaks or partial degradation. Although not frequently encountered, these events suggest that the S form is capable of provoking phagosome-cytosol communication. In conclusion, M. abscessus S exhibits traits inside macrophages that are reminiscent of slow-growing mycobacterial species.


Asunto(s)
Macrófagos/microbiología , Mycobacterium chelonae/crecimiento & desarrollo , Células Cultivadas , Transferencia Resonante de Energía de Fluorescencia , Humanos , Infecciones por Mycobacterium no Tuberculosas/microbiología , Fagosomas/microbiología
19.
Tuberculosis (Edinb) ; 95 Suppl 1: S150-4, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25732627

RESUMEN

Pathogenesis of Mycobacterium tuberculosis depends on the secretion of key virulence factors, such as the 6 kDa early secreted antigenic target ESAT-6 (EsxA) and its protein partner, the 10 kDa culture filtrate protein CFP-10 (EsxB), via the ESX-1 secretion system. ESX-1 represents the prototype system of the recently named type VII secretion systems that exist in a range of actinobacteria. The M. tuberculosis genome harbours a total of five gene clusters potentially coding for type VII secretion systems, designated ESX-1 - ESX-5, with ESX-4 being the most ancient system from which other ESX systems seem to have evolved by gene duplication and gene insertion events. The five ESX systems show similarity in gene content and gene order but differ in function. ESX-1 and ESX-5 are both crucial virulence determinants of M. tuberculosis, but with different mechanisms. While ESX-1 is implicated in the lysis of the host cell phagosomes, ESX-5 is involved in secretion of the mycobacteria specific PE and PPE proteins and cell wall stability. Research on type VII secretion systems has thus become a large and competitive research topic that is tightly linked to studies of host-pathogen interaction of pathogenic mycobacteria. Insights into this matter are of relevance for redrawing the patho-evolution of M. tuberculosis, which might help improving current strategies for prevention, diagnostics and therapy of tuberculosis as well as elucidating the virulence mechanisms employed by this important human pathogen.


Asunto(s)
Evolución Molecular , Mycobacterium tuberculosis/metabolismo , Sistemas de Secreción Tipo VII/metabolismo , Antígenos Bacterianos , Proteínas Bacterianas/metabolismo , Genoma Bacteriano/genética , Interacciones Huésped-Patógeno/genética , Humanos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Filogenia , Tuberculosis/genética , Factores de Virulencia/genética
20.
Genome Biol Evol ; 7(3): 856-70, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716827

RESUMEN

By phylogenetic analysis, Mycobacterium kansasii is closely related to Mycobacterium tuberculosis. Yet, although both organisms cause pulmonary disease, M. tuberculosis is a global health menace, whereas M. kansasii is an opportunistic pathogen. To illuminate the differences between these organisms, we have sequenced the genome of M. kansasii ATCC 12478 and its plasmid (pMK12478) and conducted side-by-side in vitro and in vivo investigations of these two organisms. The M. kansasii genome is 6,432,277 bp, more than 2 Mb longer than that of M. tuberculosis H37Rv, and the plasmid contains 144,951 bp. Pairwise comparisons reveal conserved and discordant genes and genomic regions. A notable example of genomic conservation is the virulence locus ESX-1, which is intact and functional in the low-virulence M. kansasii, potentially mediating phagosomal disruption. Differences between these organisms include a decreased predicted metabolic capacity, an increased proportion of toxin-antitoxin genes, and the acquisition of M. tuberculosis-specific genes in the pathogen since their common ancestor. Consistent with their distinct epidemiologic profiles, following infection of C57BL/6 mice, M. kansasii counts increased by less than 10-fold over 6 weeks, whereas M. tuberculosis counts increased by over 10,000-fold in just 3 weeks. Together, these data suggest that M. kansasii can serve as an image of the environmental ancestor of M. tuberculosis before its emergence as a professional pathogen, and can be used as a model organism to study the switch from an environmental opportunistic pathogen to a professional host-restricted pathogen.


Asunto(s)
Evolución Molecular , Mycobacterium kansasii/genética , Mycobacterium tuberculosis/genética , Animales , Femenino , Genoma Bacteriano , Genómica , Ratones Endogámicos C57BL , Mycobacterium kansasii/clasificación , Mycobacterium tuberculosis/clasificación , Mycobacterium tuberculosis/patogenicidad , Fenotipo , Filogenia , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA