Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS Biol ; 21(6): e3002159, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37319285

RESUMEN

The immune response to Mycobacterium tuberculosis infection determines tuberculosis disease outcomes, yet we have an incomplete understanding of what immune factors contribute to a protective immune response. Neutrophilic inflammation has been associated with poor disease prognosis in humans and in animal models during M. tuberculosis infection and, therefore, must be tightly regulated. ATG5 is an essential autophagy protein that is required in innate immune cells to control neutrophil-dominated inflammation and promote survival during M. tuberculosis infection; however, the mechanistic basis for how ATG5 regulates neutrophil recruitment is unknown. To interrogate what innate immune cells require ATG5 to control neutrophil recruitment during M. tuberculosis infection, we used different mouse strains that conditionally delete Atg5 in specific cell types. We found that ATG5 is required in CD11c+ cells (lung macrophages and dendritic cells) to control the production of proinflammatory cytokines and chemokines during M. tuberculosis infection, which would otherwise promote neutrophil recruitment. This role for ATG5 is autophagy dependent, but independent of mitophagy, LC3-associated phagocytosis, and inflammasome activation, which are the most well-characterized ways that autophagy proteins regulate inflammation. In addition to the increased proinflammatory cytokine production from macrophages during M. tuberculosis infection, loss of ATG5 in innate immune cells also results in an early induction of TH17 responses. Despite prior published in vitro cell culture experiments supporting a role for autophagy in controlling M. tuberculosis replication in macrophages, the effects of autophagy on inflammatory responses occur without changes in M. tuberculosis burden in macrophages. These findings reveal new roles for autophagy proteins in lung resident macrophages and dendritic cells that are required to suppress inflammatory responses that are associated with poor control of M. tuberculosis infection.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Ratones , Humanos , Infiltración Neutrófila , Macrófagos/fisiología , Tuberculosis/microbiología , Autofagia , Mycobacterium tuberculosis/fisiología , Inflamación
2.
Infect Immun ; 90(9): e0058821, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35972269

RESUMEN

Circadian clocks are universally used to coordinate biological processes with the Earth's 24-h solar day and are critical for the health and environmental success of an organism. Circadian rhythms in eukaryotes are driven by a cell-intrinsic transcription-translation feedback loop that controls daily oscillations in gene expression which regulate diverse physiological functions. Substantial evidence now exists demonstrating that immune activation and inflammatory responses during infection are under circadian control, however, the cellular mechanisms responsible for this are not well understood. The zebrafish (Danio rerio) is a powerful model organism to study vertebrate circadian biology and immune function. Zebrafish contain homologs of mammalian circadian clock genes which, to our current knowledge, function similarly to impart timekeeping ability. Consistent with studies in mammalian models, several studies in fish have now demonstrated a bidirectional relationship between the circadian clock and inflammation: the circadian clock regulates immune activity, and inflammation can alter circadian rhythms. This review summarizes our current understanding of the molecular mechanisms of the zebrafish clock and the bi-directional relationship between the circadian clock and inflammation in fish.


Asunto(s)
Ritmo Circadiano , Pez Cebra , Animales , Ritmo Circadiano/genética , Inflamación , Mamíferos , Pez Cebra/fisiología
3.
Front Cell Infect Microbiol ; 12: 894644, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35646729

RESUMEN

Streptococcus pneumoniae (Spn) is the primary agent of community-acquired pneumonia. Neutrophils are innate immune cells that are essential for bacterial clearance during pneumococcal pneumonia but can also do harm to host tissue. Neutrophil migration in pneumococcal pneumonia is therefore a major determinant of host disease outcomes. During Spn infection, detection of the bacterium leads to an increase in proinflammatory signals and subsequent expression of integrins and ligands on both the neutrophil as well as endothelial and epithelial cells. These integrins and ligands mediate the tethering and migration of the neutrophil from the bloodstream to the site of infection. A gradient of host-derived and bacterial-derived chemoattractants contribute to targeted movement of neutrophils. During pneumococcal pneumonia, neutrophils are rapidly recruited to the pulmonary space, but studies show that some of the canonical neutrophil migratory machinery is dispensable. Investigation of neutrophil migration is necessary for us to understand the dynamics of pneumococcal infection. Here, we summarize what is known about the pathways that lead to migration of the neutrophil from the capillaries to the lung during pneumococcal infection.


Asunto(s)
Neumonía Neumocócica , Humanos , Integrinas , Ligandos , Infiltración Neutrófila , Streptococcus pneumoniae
4.
J Innate Immun ; 13(6): 391-403, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34023827

RESUMEN

Group B Streptococcus (GBS) is a leading cause of neonatal morbidity and mortality, and the primary source of exposure is the maternal vagina. Intrapartum antibiotic prophylaxis for GBS-positive mothers has reduced the incidence of GBS early-onset disease, however, potential long-lasting influence of an antibiotic-altered neonatal microbiota, and the frequent clinical sequelae in survivors of invasive GBS infection, compels alternative treatment options for GBS. Here, we examined the role of transcription factor hypoxia-inducible factor 1 alpha (HIF-1α), widely recognized as a regulator of immune activation during infection, in the host response to GBS. Given the importance of endogenous HIF-1α for innate immune defense, and the potential utility of HIF-1α stabilization in promoting bacterial clearance, we hypothesized that HIF-1α could play an important role in coordinating host responses to GBS in colonization and systemic disease. Counter to our hypothesis, we found that GBS infection did not induce HIF-1α expression in vaginal epithelial cells or murine macrophages, nor did HIF-1α deficiency alter GBS colonization or pathogenesis in vivo. Furthermore, pharmacological enhancement of HIF-1α did not improve control of GBS in pathogenesis and colonization models, while displaying inhibitory effects in vaginal epithelial cytokines and immune cell killing in vitro. Taken together, we conclude that HIF-1α is not a prominent aspect of the host response to GBS colonization or invasive disease, and its pharmacological modulation is unlikely to provide significant benefit against this important neonatal pathogen.


Asunto(s)
Infecciones Estreptocócicas , Streptococcus agalactiae , Animales , Citocinas , Femenino , Factor 1 Inducible por Hipoxia , Ratones , Vagina
5.
Cell Host Microbe ; 29(1): 107-120.e6, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33120116

RESUMEN

Streptococcus pneumoniae is an opportunistic human pathogen that causes invasive diseases, including pneumonia, with greater health risks upon influenza A virus (IAV) co-infection. To facilitate pathogenesis studies in vivo, we developed an inducible CRISPR interference system that enables genome-wide fitness testing in one sequencing step (CRISPRi-seq). We applied CRISPRi-seq to assess bottlenecks and identify pneumococcal genes important in a murine pneumonia model. A critical bottleneck occurs at 48 h with few bacteria causing systemic infection. This bottleneck is not present during IAV superinfection, facilitating identification of pneumococcal pathogenesis-related genes. Top in vivo essential genes included purA, encoding adenylsuccinate synthetase, and the cps operon required for capsule production. Surprisingly, CRISPRi-seq indicated no fitness-related role for pneumolysin during superinfection. Interestingly, although metK (encoding S-adenosylmethionine synthetase) was essential in vitro, it was dispensable in vivo. This highlights advantages of CRISPRi-seq over transposon-based genetic screens, as all genes, including essential genes, can be tested for pathogenesis potential.


Asunto(s)
Genes Bacterianos , Neumonía Neumocócica/microbiología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Adenilosuccinato Sintasa/genética , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Femenino , Aptitud Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Virus de la Influenza A , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Operón , Infecciones por Orthomyxoviridae/complicaciones , Neumonía Neumocócica/complicaciones , Streptococcus pneumoniae/crecimiento & desarrollo , Sobreinfección
6.
Sci Rep ; 10(1): 21738, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33303951

RESUMEN

Serological testing to evaluate antigen-specific antibodies in plasma is generally performed by rapid lateral flow test strips that lack quantitative results or by high complexity immunoassays that are time- and labor-intensive but provide semi-quantitative results. Here, we describe a novel application of biolayer interferometry for the rapid detection of antigen-specific antibody levels in plasma samples, and demonstrate its utility for quantification of SARS-CoV-2 antibodies. Our biolayer interferometry immunosorbent assay (BLI-ISA) utilizes single-use biosensors in an automated "dip-and-read" format, providing real-time optical measurements of antigen loading, plasma antibody binding, and antibody isotype detection. Complete semi-quantitative results are obtained in less than 20 min. BLI-ISA meets or exceeds the performance of high complexity methods such as Enzyme-Linked Immunosorbent Assay (ELISA) and Chemiluminescent Immunoassay. Importantly, our method can be immediately implemented on existing BLI platforms for urgent COVID-19 studies, such as serosurveillance and the evaluation of vaccine candidates. In a broader sense, BLI-ISA can be developed as a novel diagnostic platform to evaluate antibodies and other biomolecules in clinical specimens.


Asunto(s)
Anticuerpos Antivirales , Prueba Serológica para COVID-19 , COVID-19 , SARS-CoV-2 , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , COVID-19/sangre , COVID-19/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Interferometría , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo
7.
EBioMedicine ; 60: 102984, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32979835

RESUMEN

BACKGROUND: Pulmonary damage by Pseudomonas aeruginosa during cystic fibrosis lung infection and ventilator-associated pneumonia is mediated both by pathogen virulence factors and host inflammation. Impaired immune function due to tissue damage and inflammation, coupled with pathogen multidrug resistance, complicates the management of these deep-seated infections. Pathological inflammation during infection is driven by interleukin-1ß (IL-1ß), but the molecular processes involved are not fully understood. METHODS: We examined IL-1ß activation in a pulmonary model infection of Pseudomonas aeruginosa and in vitro using genetics, specific inhibitors, recombinant proteins, and targeted reporters of protease activity and IL-1ß bioactivity. FINDINGS: Caspase-family inflammasome proteases canonically regulate maturation of this proinflammatory cytokine, but we report that plasticity in IL-1ß proteolytic activation allows for its direct maturation by the pseudomonal protease LasB. LasB promotes IL-1ß activation, neutrophilic inflammation, and destruction of lung architecture characteristic of severe P. aeruginosa pulmonary infection. INTERPRETATION: Preservation of lung function and effective immune clearance may be enhanced by selectively controlling inflammation. Discovery of this IL-1ß regulatory mechanism provides a distinct target for anti-inflammatory therapeutics, such as matrix metalloprotease inhibitors that inhibit LasB and limit inflammation and pathology during P. aeruginosa pulmonary infections. FUNDING: Full details are provided in the Acknowledgements section.


Asunto(s)
Interacciones Huésped-Patógeno , Interleucina-1beta/metabolismo , Pseudomonas aeruginosa/enzimología , Serina Endopeptidasas/metabolismo , Animales , Biomarcadores , Fibrosis Quística/complicaciones , Fibrosis Quística/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Inmunohistoquímica , Inflamasomas/metabolismo , Mediadores de Inflamación , Metaloproteasas/antagonistas & inhibidores , Ratones , Ratones Noqueados , Modelos Biológicos , Neumonía Bacteriana/etiología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología , Unión Proteica , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología
8.
medRxiv ; 2020 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-32743612

RESUMEN

Serological testing to evaluate antigen-specific antibodies in plasma is generally performed by rapid lateral flow test strips that lack quantitative results or by high complexity immunoassays that are time- and labor-intensive but provide quantitative results. Here, we describe a novel application of biolayer interferometry for the rapid detection of antigen-specific antibody levels in plasma samples, and demonstrate its utility for quantification of SARS-CoV-2 antibodies. Our biolayer interferometry immunosorbent assay (BLI-ISA) utilizes single-use biosensors in an automated "dip-and-read" format, providing real-time optical measurements of antigen loading, plasma antibody binding, and antibody isotype detection. Complete quantitative results are obtained in less than 20 minutes. BLI-ISA meets or exceeds the performance of high complexity methods such as Enzyme-Linked Immunosorbent Assay (ELISA) and Chemiluminescent Immunoassay. Importantly, our method can be immediately implemented on existing BLI platforms for urgent COVID-19 studies, such as serosurveillance and the evaluation of vaccine candidates. In a broader sense, BLI-ISA can be developed as a novel diagnostic platform to evaluate antibodies and other biomolecules in clinical specimens.

9.
iScience ; 23(6): 101207, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32535023

RESUMEN

Streptococcus agalactiae (Group B Streptococcus, GBS) is the most common neonatal pathogen. However, the cellular and molecular mechanisms for neonatal susceptibility to GBS pneumonia and sepsis are incompletely understood. Here we optimized a mouse model of GBS pneumonia to test the role of alveolar macrophage (ΑΜΦ) maturation in host vulnerability to disease. Compared with juvenile and adult mice, neonatal mice infected with GBS had increased mortality and persistence of lung injury. In addition, neonatal mice were defective in GBS phagocytosis and killing. ΑΜΦ depletion and disruption of ΑΜΦ differentiation in Csf2-/- mice both impaired GBS clearance. AMΦ engage the heavily sialylated GBS capsule via the cell surface Siglec receptors Sn and Siglec-E. Although both newborn and adult ΑΜΦ expressed Siglec-E, newborn ΑΜΦ expressed significantly lower levels of Sn. We propose that a developmental delay in Sn expression on ΑΜΦ may prevent effective killing and clearing of GBS from the newborn lung.

10.
J Exp Med ; 215(7): 1823-1838, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29773644

RESUMEN

The cytokine IL-10 antagonizes pathways that control Mycobacterium tuberculosis (Mtb) infection. Nevertheless, the impact of IL-10 during Mtb infection has been difficult to decipher because loss-of-function studies in animal models have yielded only mild phenotypes. We have discovered that the transcription factor basic helix-loop-helix family member e40 (Bhlhe40) is required to repress Il10 expression during Mtb infection. Loss of Bhlhe40 in mice results in higher Il10 expression, higher bacterial burden, and early susceptibility similar to that observed in mice lacking IFN-γ. Deletion of Il10 in Bhlhe40-/- mice reverses these phenotypes. Bhlhe40 deletion in T cells or CD11c+ cells is sufficient to cause susceptibility to Mtb Bhlhe40 represents the first transcription factor found to be essential during Mtb infection to specifically regulate Il10 expression, revealing the importance of strict control of IL-10 production by innate and adaptive immune cells during infection. Our findings uncover a previously elusive but significant role for IL-10 in Mtb pathogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Homeodominio/metabolismo , Interleucina-10/metabolismo , Proteínas Represoras/metabolismo , Tuberculosis/inmunología , Inmunidad Adaptativa , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Sitios Genéticos , Inmunidad Innata , Inflamación/patología , Interleucina-10/deficiencia , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Células Mieloides/metabolismo , Neutrófilos/metabolismo , Unión Proteica , Células TH1/metabolismo , Tuberculosis/prevención & control
11.
J Biol Chem ; 293(16): 6022-6038, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29496999

RESUMEN

Germline-encoded receptors recognizing common pathogen-associated molecular patterns are a central element of the innate immune system and play an important role in shaping the host response to infection. Many of the innate immune molecules central to these signaling pathways are evolutionarily conserved. LysMD3 is a novel molecule containing a putative peptidoglycan-binding domain that has orthologs in humans, mice, zebrafish, flies, and worms. We found that the lysin motif (LysM) of LysMD3 is likely related to a previously described peptidoglycan-binding LysM found in bacteria. Mouse LysMD3 is a type II integral membrane protein that co-localizes with GM130+ structures, consistent with localization to the Golgi apparatus. We describe here two lines of mLysMD3-deficient mice for in vivo characterization of mLysMD3 function. We found that mLysMD3-deficient mice were born at Mendelian ratios and had no obvious pathological abnormalities. They also exhibited no obvious immune response deficiencies in a number of models of infection and inflammation. mLysMD3-deficient mice exhibited no signs of intestinal dysbiosis by 16S analysis or alterations in intestinal gene expression by RNA sequencing. We conclude that mLysMD3 contains a LysM with cytoplasmic orientation, but we were unable to define a physiological role for the molecule in vivo.


Asunto(s)
Eliminación de Gen , Animales , Autoantígenos/análisis , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Sistemas CRISPR-Cas , Femenino , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Masculino , Proteínas de la Membrana/análisis , Ratones , Micosis/genética , Micosis/inmunología , Filogenia , Virosis/genética , Virosis/inmunología
12.
PLoS Genet ; 13(11): e1007115, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29176877

RESUMEN

DNA replication is fundamental for life, yet a detailed understanding of bacterial DNA replication is limited outside the organisms Escherichia coli and Bacillus subtilis. Many bacteria, including mycobacteria, encode no identified homologs of helicase loaders or regulators of the initiator protein DnaA, despite these factors being essential for DNA replication in E. coli and B. subtilis. In this study we discover that a previously uncharacterized protein, Rv0004, from the human pathogen Mycobacterium tuberculosis is essential for bacterial viability and that depletion of Rv0004 leads to a block in cell cycle progression. Using a combination of genetic and biochemical approaches, we found that Rv0004 has a role in DNA replication, interacts with DNA and the replicative helicase DnaB, and affects DnaB-DnaA complex formation. We also identify a conserved domain in Rv0004 that is predicted to structurally resemble the N-terminal protein-protein interaction domain of DnaA. Mutation of a single conserved tryptophan within Rv0004's DnaA N-terminal-like domain leads to phenotypes similar to those observed upon Rv0004 depletion and can affect the association of Rv0004 with DnaB. In addition, using live cell imaging during depletion of Rv0004, we have uncovered a previously unappreciated role for DNA replication in coordinating mycobacterial cell division and cell size. Together, our data support that Rv0004 encodes a homolog of the recently identified DciA family of proteins found in most bacteria that lack the DnaC-DnaI helicase loaders in E. coli and B. subtilis. Therefore, the mechanisms of Rv0004 elucidated here likely apply to other DciA homologs and reveal insight into the diversity of bacterial strategies in even the most conserved biological processes.


Asunto(s)
Proteínas Bacterianas/genética , Replicación del ADN/genética , ADN Bacteriano/genética , Proteínas de Unión al ADN/genética , Mycobacterium tuberculosis/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Ciclo Celular/genética , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , AdnB Helicasas/metabolismo , Viabilidad Microbiana/genética , Mycobacterium tuberculosis/metabolismo , Unión Proteica , Homología de Secuencia de Aminoácido
13.
Microbes Infect ; 19(4-5): 249-258, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28087453

RESUMEN

Mycobacterium tuberculosis infection results in 1.5 million deaths annually. Type I interferon (IFN) signaling through its receptor IFNAR correlates with increased severity of disease, although how this increases susceptibility to M. tuberculosis remains uncertain. ISG15 is one of the most highly induced interferon stimulated genes (ISGs) during M. tuberculosis infection. ISG15 functions by conjugation to target proteins (ISGylation), by noncovalent association with intracellular proteins, and by release from the cell. Recent studies indicated that ISG15 can function via conjugation-independent mechanisms to suppress the type I IFN response. These data raised the question of whether ISG15 may have diverse and sometimes opposing functions during M. tuberculosis infection. To address this, we analyzed ISGylation during M. tuberculosis infection and show that ISGylated proteins accumulate following infection in an IFNAR-dependent manner. Type I IFN and ISG15 both play transient roles in promoting bacterial replication. However, as the disease progresses, ISGylation deviates from the overall effect of type I IFN and, ultimately, mice deficient in ISGylation are significantly more susceptible than IFNAR mice. Our data demonstrate that ISGs can both protect against and promote disease and are the first to report a role for ISGylation during M. tuberculosis infection.


Asunto(s)
Citocinas/genética , Interferón Tipo I/inmunología , Mycobacterium tuberculosis/inmunología , Receptor de Interferón alfa y beta/genética , Tuberculosis Pulmonar/patología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/metabolismo , Unión Proteica/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Tuberculosis Pulmonar/microbiología , Ubiquitinas/genética
14.
Trends Mol Med ; 22(12): 1060-1076, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27866924

RESUMEN

Research in recent years has focused significantly on the role of selective macroautophagy in targeting intracellular pathogens for lysosomal degradation, a process termed xenophagy. In this review we evaluate the proposed roles for xenophagy in controlling bacterial infection, highlighting the concept that successful pathogens have evolved ways to subvert or exploit this defense, minimizing the actual effectiveness of xenophagy in innate immunity. Instead, studies in animal models have revealed that autophagy-associated proteins often function outside of xenophagy to influence bacterial pathogenesis. In light of current efforts to manipulate autophagy and the development of host-directed therapies to fight bacterial infections, we also discuss the implications stemming from the complicated relationship that exists between autophagy and bacterial pathogens.


Asunto(s)
Autofagia , Bacterias/inmunología , Infecciones Bacterianas/inmunología , Inmunidad Innata , Animales , Antibacterianos/farmacología , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/inmunología , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/patología , Fenómenos Fisiológicos Bacterianos , Descubrimiento de Drogas , Interacciones Huésped-Patógeno , Humanos , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/fisiología , Tuberculosis/tratamiento farmacológico , Tuberculosis/inmunología , Tuberculosis/patología
15.
Microbiology (Reading) ; 162(10): 1784-1796, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27586540

RESUMEN

Mycobacterium tuberculosis (Mtb) is one of the world's most successful pathogens. Millions of new cases of tuberculosis occur each year, emphasizing the need for better methods of treatment. The design of novel therapeutics is dependent on our understanding of factors that are essential for pathogenesis. Many bacterial pathogens use pili and other adhesins to mediate pathogenesis. The recently identified Mycobacterium tuberculosis pilus (MTP) and the hypothetical, widely conserved Flp pilus have been speculated to be important for Mtb virulence based on in vitro studies and homology to other pili, respectively. However, the roles for these pili during infection have yet to be tested. We addressed this gap in knowledge and found that neither MTP nor the hypothetical Flp pilus is required for Mtb survival in mouse models of infection, although MTP can contribute to biofilm formation and subsequent isoniazid tolerance. However, differences in mtp expression did affect lesion architecture in infected lungs. Deletion of mtp did not correlate with loss of cell-associated extracellular structures as visualized by transmission electron microscopy in Mtb Erdman and HN878 strains, suggesting that the phenotypes of the mtp mutants were not due to defects in production of extracellular structures. These findings highlight the importance of testing the virulence of adhesion mutants in animal models to assess the contribution of the adhesin to infection. This study also underscores the need for further investigation into additional strategies that Mtb may use to adhere to its host so that we may understand how this pathogen invades, colonizes and disseminates.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fimbrias Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/microbiología , Animales , Proteínas Bacterianas/genética , Femenino , Fimbrias Bacterianas/genética , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/genética , Virulencia
16.
Nature ; 528(7583): 565-9, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26649827

RESUMEN

Mycobacterium tuberculosis, a major global health threat, replicates in macrophages in part by inhibiting phagosome-lysosome fusion, until interferon-γ (IFNγ) activates the macrophage to traffic M. tuberculosis to the lysosome. How IFNγ elicits this effect is unknown, but many studies suggest a role for macroautophagy (herein termed autophagy), a process by which cytoplasmic contents are targeted for lysosomal degradation. The involvement of autophagy has been defined based on studies in cultured cells where M. tuberculosis co-localizes with autophagy factors ATG5, ATG12, ATG16L1, p62, NDP52, BECN1 and LC3 (refs 2-6), stimulation of autophagy increases bacterial killing, and inhibition of autophagy increases bacterial survival. Notably, these studies reveal modest (~1.5-3-fold change) effects on M. tuberculosis replication. By contrast, mice lacking ATG5 in monocyte-derived cells and neutrophils (polymorponuclear cells, PMNs) succumb to M. tuberculosis within 30 days, an extremely severe phenotype similar to mice lacking IFNγ signalling. Importantly, ATG5 is the only autophagy factor that has been studied during M. tuberculosis infection in vivo and autophagy-independent functions of ATG5 have been described. For this reason, we used a genetic approach to elucidate the role for multiple autophagy-related genes and the requirement for autophagy in resistance to M. tuberculosis infection in vivo. Here we show that, contrary to expectation, autophagic capacity does not correlate with the outcome of M. tuberculosis infection. Instead, ATG5 plays a unique role in protection against M. tuberculosis by preventing PMN-mediated immunopathology. Furthermore, while Atg5 is dispensable in alveolar macrophages during M. tuberculosis infection, loss of Atg5 in PMNs can sensitize mice to M. tuberculosis. These findings shift our understanding of the role of ATG5 during M. tuberculosis infection, reveal new outcomes of ATG5 activity, and shed light on early events in innate immunity that are required to regulate disease pathology and bacterial replication.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Mycobacterium tuberculosis , Neutrófilos/inmunología , Tuberculosis/inmunología , Tuberculosis/patología , Animales , Autofagia/genética , Proteína 5 Relacionada con la Autofagia , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Inmunidad Innata/inmunología , Interferón gamma/deficiencia , Interferón gamma/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/fisiología , Neutrófilos/metabolismo , Tuberculosis/microbiología
17.
Cell Host Microbe ; 17(6): 811-819, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26048136

RESUMEN

Type I interferons (IFNs) are critical mediators of antiviral defense, but their elicitation by bacterial pathogens can be detrimental to hosts. Many intracellular bacterial pathogens, including Mycobacterium tuberculosis, induce type I IFNs following phagosomal membrane perturbations. Cytosolic M. tuberculosis DNA has been implicated as a trigger for IFN production, but the mechanisms remain obscure. We report that the cytosolic DNA sensor, cyclic GMP-AMP synthase (cGAS), is required for activating IFN production via the STING/TBK1/IRF3 pathway during M. tuberculosis and L. pneumophila infection of macrophages, whereas L. monocytogenes short-circuits this pathway by producing the STING agonist, c-di-AMP. Upon sensing cytosolic DNA, cGAS also activates cell-intrinsic antibacterial defenses, promoting autophagic targeting of M. tuberculosis. Importantly, we show that cGAS binds M. tuberculosis DNA during infection, providing direct evidence that this unique host-pathogen interaction occurs in vivo. These data uncover a mechanism by which IFN is likely elicited during active human infections.


Asunto(s)
ADN Bacteriano/metabolismo , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Mycobacterium tuberculosis/genética , Nucleotidiltransferasas/metabolismo , Animales , Antígenos Bacterianos/metabolismo , Autofagia/fisiología , Proteínas Bacterianas/metabolismo , Citosol/metabolismo , Femenino , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Mycobacterium tuberculosis/patogenicidad , Nucleotidiltransferasas/genética , Tuberculosis/microbiología
18.
Elife ; 42015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25599590

RESUMEN

Variation in the presentation of hereditary immunodeficiencies may be explained by genetic or environmental factors. Patients with mutations in HOIL1 (RBCK1) present with amylopectinosis-associated myopathy with or without hyper-inflammation and immunodeficiency. We report that barrier-raised HOIL-1-deficient mice exhibit amylopectin-like deposits in the myocardium but show minimal signs of hyper-inflammation. However, they show immunodeficiency upon acute infection with Listeria monocytogenes, Toxoplasma gondii or Citrobacter rodentium. Increased susceptibility to Listeria was due to HOIL-1 function in hematopoietic cells and macrophages in production of protective cytokines. In contrast, HOIL-1-deficient mice showed enhanced control of chronic Mycobacterium tuberculosis or murine γ-herpesvirus 68 (MHV68), and these infections conferred a hyper-inflammatory phenotype. Surprisingly, chronic infection with MHV68 complemented the immunodeficiency of HOIL-1, IL-6, Caspase-1 and Caspase-1;Caspase-11-deficient mice following Listeria infection. Thus chronic herpesvirus infection generates signs of auto-inflammation and complements genetic immunodeficiency in mutant mice, highlighting the importance of accounting for the virome in genotype-phenotype studies.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Herpesviridae/fisiología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/virología , Enfermedad Aguda , Animales , Células de la Médula Ósea/citología , Caspasa 1/metabolismo , Compartimento Celular , Enfermedad Crónica , Citrobacter/fisiología , Citocinas/biosíntesis , Prueba de Complementación Genética , Infecciones por Herpesviridae/virología , Humanos , Inmunidad Innata , Inflamación/patología , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Listeriosis/microbiología , Listeriosis/patología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/fisiología , Fenotipo , Rhadinovirus/fisiología , Toxoplasma , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(12): 5051-6, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21383189

RESUMEN

Mycobacterium tuberculosis must import iron from its host for survival, and its siderophore-dependent iron acquisition pathways are well established. Here we demonstrate a newly characterized pathway, whereby M. tuberculosis can use free heme and heme from hemoglobin as an iron source. Significantly, we identified the genomic region, Rv0202c-Rv0207c, responsible for the passage of heme iron across the mycobacterial membrane. Key players of this heme uptake system were characterized including a secreted protein and two transmembrane proteins, all three specific to mycobacteria. Furthermore, the crystal structure of the key heme carrier protein Rv0203 was found to have a unique fold. The discovery of a unique mycobacterial heme acquisition pathway opens new avenues of exploration into mycobacterial therapeutics.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Hemo/metabolismo , Hierro/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteínas Bacterianas/genética , Transporte Biológico/fisiología , Proteínas Portadoras/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Hemo/genética , Mycobacterium tuberculosis/genética , Tuberculosis/tratamiento farmacológico , Tuberculosis/genética , Tuberculosis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA