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













Base de datos
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.
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
4.
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.

5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
J Vis Exp ; (76): e50116, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23792688

RESUMEN

Shigella flexneri are pathogenic bacteria that invade host cells entering into an endocytic vacuole. Subsequently, the rupture of this membrane-enclosed compartment allows bacteria to move within the cytosol, proliferate and further invade neighboring cells. Mycobacterium tuberculosis is phagocytosed by immune cells, and has recently been shown to rupture phagosomal membrane in macrophages. We developed a robust assay for tracking phagosomal membrane disruption after host cell entry of Shigella flexneri or Mycobacterium tuberculosis. The approach makes use of CCF4, a FRET reporter sensitive to ß-lactamase that equilibrates in the cytosol of host cells. Upon invasion of host cells by bacterial pathogens, the probe remains intact as long as the bacteria reside in membrane-enclosed compartments. After disruption of the vacuole, ß-lactamase activity on the surface of the intracellular pathogen cleaves CCF4 instantly leading to a loss of FRET signal and switching its emission spectrum. This robust ratiometric assay yields accurate information about the timing of vacuolar rupture induced by the invading bacteria, and it can be coupled to automated microscopy and image processing by specialized algorithms for the detection of the emission signals of the FRET donor and acceptor. Further, it allows investigating the dynamics of vacuolar disruption elicited by intracellular bacteria in real time in single cells. Finally, it is perfectly suited for high-throughput analysis with a spatio-temporal resolution exceeding previous methods. Here, we provide the experimental details of exemplary protocols for the CCF4 vacuolar rupture assay on HeLa cells and THP-1 macrophages for time-lapse experiments or end points experiments using Shigella flexneri as well as multiple mycobacterial strains such as Mycobacterium marinum, Mycobacterium bovis, and Mycobacterium tuberculosis.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Shigella flexneri/patogenicidad , Vacuolas/microbiología , Línea Celular , Disentería Bacilar/metabolismo , Disentería Bacilar/microbiología , Disentería Bacilar/patología , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Humanos , Membranas Intracelulares , Macrófagos/microbiología , Shigella flexneri/enzimología , Vacuolas/metabolismo , Vacuolas/patología , beta-Lactamasas/metabolismo
14.
PLoS One ; 8(3): e58954, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23536839

RESUMEN

Phenolic glycolipids are produced by a very limited number of slow-growing mycobacterial species, most of which are pathogen for humans. In Mycobacterium tuberculosis, the etiologic agent of tuberculosis, these molecules play a role in the pathogenicity by modulating the host immune response during infection. The major variant of phenolic glycolipids produced by M. tuberculosis, named PGL-tb, consists of a large lipid core terminated by a glycosylated aromatic nucleus. The carbohydrate part is composed of three sugar residues, two rhamnosyl units and a terminal fucosyl residue, which is per-O-methylated, and seems to be important for pathogenicity. While most of the genes responsible for the synthesis of the lipid core domain and the saccharide appendage of PGL-tb have been characterized, the enzymes involved in the O-methylation of the fucosyl residue of PGL-tb remain unknown. In this study we report the identification and characterization of the methyltransferases required for the O-methylation of the terminal fucosyl residue of PGL-tb. These enzymes are encoded by genes Rv2954c, Rv2955c and Rv2956. Mutants of M. tuberculosis harboring deletion within these genes were constructed. Purification and analysis of the phenolglycolipids produced by these strains, using a combination of mass spectrometry and NMR spectroscopy, revealed that Rv2954c, Rv2955c and Rv2956 encode the methyltransferases that respectively catalysed the O-methylation of the hydroxyl groups located at positions 3, 4 and 2 of the terminal fucosyl residue of PGL-tb. Our data also suggest that methylation at these positions is a sequential process, starting with position 2, followed by positions 4 and 3.


Asunto(s)
Glucolípidos/biosíntesis , Metiltransferasas/metabolismo , Mycobacterium tuberculosis/metabolismo , Secuencia de Aminoácidos , Animales , Catálisis , Orden Génico , Glucolípidos/química , Humanos , Metilación , Metiltransferasas/química , Metiltransferasas/genética , Datos de Secuencia Molecular , Mutación , Mycobacterium tuberculosis/genética , Alineación de Secuencia
15.
Nat Genet ; 45(2): 172-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23291586

RESUMEN

Global spread and limited genetic variation are hallmarks of M. tuberculosis, the agent of human tuberculosis. In contrast, Mycobacterium canettii and related tubercle bacilli that also cause human tuberculosis and exhibit unusual smooth colony morphology are restricted to East Africa. Here, we sequenced and analyzed the whole genomes of five representative strains of smooth tubercle bacilli (STB) using Sanger (4-5× coverage), 454/Roche (13-18× coverage) and/or Illumina DNA sequencing (45-105× coverage). We show that STB isolates are highly recombinogenic and evolutionarily early branching, with larger genome sizes, higher rates of genetic variation, fewer molecular scars and distinct CRISPR-Cas systems relative to M. tuberculosis. Despite the differences, all tuberculosis-causing mycobacteria share a highly conserved core genome. Mouse infection experiments showed that STB strains are less persistent and virulent than M. tuberculosis. We conclude that M. tuberculosis emerged from an ancestral STB-like pool of mycobacteria by gain of persistence and virulence mechanisms, and we provide insights into the molecular events involved.


Asunto(s)
Adaptación Biológica/genética , Adaptación Biológica/inmunología , Evolución Molecular , Variación Genética , Genoma Bacteriano/genética , Mycobacterium tuberculosis/genética , Filogenia , Adaptación Biológica/fisiología , Animales , Secuencia de Bases , Análisis por Conglomerados , Genómica , Secuencias Invertidas Repetidas/genética , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Mycobacterium tuberculosis/clasificación , Mycobacterium tuberculosis/patogenicidad , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ADN , Especificidad de la Especie , Bazo/virología , Virulencia
16.
Autophagy ; 8(9): 1357-70, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22885411

RESUMEN

Emerging evidence points to an important role of autophagy in the immune response mediated by dendritic cells (DC) against Mycobacterium tuberculosis (Mtb). Since current vaccination based on Bacillus Calmette-Guerin (BCG) is unable to stop the tuberculosis epidemic, a deeper comprehension of the alterations induced by Mtb in DC is essential for setting new vaccine strategies. Here, we compared the capacity of virulent (H37Rv) and avirulent (H37Ra) Mtb strains as well as BCG to modulate autophagy in human primary DC. We found that Mtb H37Rv impairs autophagy at the step of autophagosome-lysosome fusion. In contrast, neither Mtb H37Ra nor BCG strains were able to hamper autophagosome maturation. Both these attenuated strains have a functional inhibition of the 6kD early secreted antigenic target ESAT-6, an effector protein of the ESAT-6 Secretion System-1(ESX-1)/type VII secretion system. Notably, the ability to inhibit autophagy was fully restored in recombinant BCG and Mtb H37Ra strains in which ESAT-6 secretion was re-established by genetic complementation using either the ESX-1 region from Mtb (BCG::ESX-1) or the PhoP gene (Mtb H37Ra::PhoP), a regulator of ESAT-6 secretion. Importantly, the autophagic block induced by Mtb was overcome by rapamycin treatment leading to an increased interleukin-12 expression and, in turn, to an enhanced capacity to expand a Th1-oriented response. Collectively, our study demonstrated that Mtb alters the autophagic machinery through the ESX-1 system, and thereby opens new exciting perspectives to better understand the relationship between Mtb virulence and its ability to escape the DC-mediated immune response.


Asunto(s)
Autofagia , Sistemas de Secreción Bacterianos , Células Dendríticas/microbiología , Células Dendríticas/patología , Mycobacterium tuberculosis/fisiología , Autofagia/efectos de los fármacos , Sistemas de Secreción Bacterianos/efectos de los fármacos , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/ultraestructura , Humanos , Mycobacterium bovis/efectos de los fármacos , Mycobacterium bovis/fisiología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/ultraestructura , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Sirolimus/farmacología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis/patología
17.
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
18.
Cell Host Microbe ; 11(4): 352-63, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-22520463

RESUMEN

The genome of Mycobacterium tuberculosis (Mtb) encodes five type VII secretion systems, ESX-1 to ESX-5, most of which are associated with genes encoding PE/PPE proteins, named after their N-terminal Pro-Glu (PE) or Pro-Pro-Glu (PPE) motifs. Here, we describe the strong T cell immunogenicity of the ESX-5-encoded PE/PPE proteins, which share a large panel of cross-reactive CD4(+) epitopes with substantial numbers of their ESX-5-nonassociated PE/PPE homologs. The immunogenicity of these numerous PE/PPE proteins is dependent on their export by a functional EccD(5), the predicted transmembrane channel of the ESX-5 secretion apparatus. The Mtb Δppe25-pe19 mutant deleted for all ESX-5-associated pe and ppe genes, although highly attenuated in immunocompetent mice, remains able to induce immunity against the ESX-5-associated PE/PPE virulence factors, via cross-reactivity with their numerous homologs, and against the ESX-1 virulence factors ESAT-6/CFP-10. The Δppe25-pe19 strain is strongly protective against Mtb infection in mice and represents a potential antituberculosis vaccine candidate.


Asunto(s)
Proteínas Bacterianas/inmunología , Sistemas de Secreción Bacterianos , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/microbiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/inmunología , Variación Antigénica , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Reacciones Cruzadas , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Datos de Secuencia Molecular , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Transporte de Proteínas , Tuberculosis/inmunología , Vacunas contra la Tuberculosis/química , Vacunas contra la Tuberculosis/genética
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA