Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 21(11): e50830, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33124745

RESUMEN

Inflammation associated with gram-negative bacterial infections is often instigated by the bacterial cell wall component lipopolysaccharide (LPS). LPS-induced inflammation and resulting life-threatening sepsis are mediated by the two distinct LPS receptors TLR4 and caspase-11 (caspase-4/-5 in humans). Whereas the regulation of TLR4 activation by extracellular and phago-endosomal LPS has been studied in great detail, auxiliary host factors that specifically modulate recognition of cytosolic LPS by caspase-11 are largely unknown. This study identifies autophagy-related and dynamin-related membrane remodeling proteins belonging to the family of Immunity-related GTPases M clade (IRGM) as negative regulators of caspase-11 activation in macrophages. Phagocytes lacking expression of mouse isoform Irgm2 aberrantly activate caspase-11-dependent inflammatory responses when exposed to extracellular LPS, bacterial outer membrane vesicles, or gram-negative bacteria. Consequently, Irgm2-deficient mice display increased susceptibility to caspase-11-mediated septic shock in vivo. This Irgm2 phenotype is partly reversed by the simultaneous genetic deletion of the two additional Irgm paralogs Irgm1 and Irgm3, indicating that dysregulated Irgm isoform expression disrupts intracellular LPS processing pathways that limit LPS availability for caspase-11 activation.


Asunto(s)
Lipopolisacáridos , Choque Séptico , Animales , Caspasas/genética , Caspasas Iniciadoras , Dinaminas , Inflamasomas , Lipopolisacáridos/toxicidad , Ratones , Choque Séptico/inducido químicamente , Choque Séptico/genética
2.
mBio ; 8(5)2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974614

RESUMEN

The Gram-negative bacterial cell wall component lipopolysaccharide (LPS) is recognized by the noncanonical inflammasome protein caspase-11 in the cytosol of infected host cells and thereby prompts an inflammatory immune response linked to sepsis. Host guanylate binding proteins (GBPs) promote infection-induced caspase-11 activation in tissue culture models, and yet their in vivo role in LPS-mediated sepsis has remained unexplored. LPS can be released from lysed bacteria as "free" LPS aggregates or actively secreted by live bacteria as a component of outer membrane vesicles (OMVs). Here, we report that GBPs control inflammation and sepsis in mice injected with either free LPS or purified OMVs derived from Gram-negative Escherichia coli In agreement with our observations from in vivo experiments, we demonstrate that macrophages lacking GBP2 expression fail to induce pyroptotic cell death and proinflammatory interleukin-1ß (IL-1ß) and IL-18 secretion when exposed to OMVs. We propose that in order to activate caspase-11 in vivo, GBPs control the processing of bacterium-derived OMVs by macrophages as well as the processing of circulating free LPS by as-yet-undetermined cell types.IMPORTANCE The bacterial cell wall component LPS is a strong inducer of inflammation and is responsible for much of the toxicity of Gram-negative bacteria. Bacteria shed some of their cell wall and its associated LPS in the form of outer membrane vesicles (OMVs). Recent work demonstrated that secreted OMVs deliver LPS into the host cell cytosol by an unknown mechanism, resulting in the activation of the proinflammatory LPS sensor caspase-11. Here, we show that activation of cytosolic caspase-11 by OMVs requires additional host factors, the so-called guanylate binding proteins (GBPs). The discovery of GBPs as regulators of OMV-mediated inflammation paves the way toward a mechanistic understanding of the host response toward bacterial OMVs and may lead to effective strategies to ameliorate inflammation induced by bacterial infections.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Escherichia coli/patogenicidad , Proteínas de Unión al GTP/metabolismo , Inflamasomas/inmunología , Inflamasomas/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Animales , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/genética , Caspasas/metabolismo , Caspasas Iniciadoras , Células Cultivadas , Citosol/metabolismo , Activación Enzimática , Inflamación , Interleucina-18/biosíntesis , Interleucina-1beta/biosíntesis , Lipopolisacáridos/inmunología , Ratones , Piroptosis , Vesículas Secretoras/metabolismo
3.
mBio ; 7(6)2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27965446

RESUMEN

The cytokine gamma interferon (IFN-γ) induces cell-autonomous immunity to combat infections with intracellular pathogens, such as the bacterium Chlamydia trachomatis The present study demonstrates that IFN-γ-primed human cells ubiquitinate and eliminate intracellular Chlamydia-containing vacuoles, so-called inclusions. We previously described how IFN-γ-inducible immunity-related GTPases (IRGs) employ ubiquitin systems to mark inclusions for destruction in mouse cells and, furthermore, showed that the rodent pathogen Chlamydia muridarum blocks ubiquitination of its inclusions by interfering with mouse IRG function. Here, we report that ubiquitination of inclusions in human cells is independent of IRG and thus distinct from the murine pathway. We show that C. muridarum is susceptible to inclusion ubiquitination in human cells, while the closely related human pathogen C. trachomatis is resistant. C. muridarum, but not C. trachomatis, inclusions attract several markers of cell-autonomous immunity, including the ubiquitin-binding protein p62, the ubiquitin-like protein LC3, and guanylate-binding protein 1. Consequently, we find that IFN-γ priming of human epithelial cells triggers the elimination of C. muridarum, but not C. trachomatis, inclusions. This newly described defense pathway is independent of indole-2,3-dioxygenase, a known IFN-γ-inducible anti-Chlamydia resistance factor. Collectively, our observations indicate that C. trachomatis evolved mechanisms to avoid a human-specific, ubiquitin-mediated response as part of its unique adaptation to its human host. IMPORTANCE: Chlamydia trachomatis is the leading cause of sexually transmitted bacterial infections and responsible for significant morbidity, including pelvic inflammatory disease, infertility, and ectopic pregnancies in women. As an obligate intracellular pathogen, C. trachomatis is in perpetual conflict with cell-intrinsic defense programs executed by its human host. Our study defines a novel anti-Chlamydia host resistance pathway active in human epithelial cells. This defense program promotes the deposition of the small antimicrobial protein ubiquitin on vacuoles containing Chlamydia We show that this ubiquitin-based resistance pathway of human cells is highly effective against a Chlamydia species adapted to rodents but ineffective against human-adapted C. trachomatis This observation indicates that C. trachomatis evolved strategies to avoid entrapment within ubiquitin-labeled vacuoles as part of its adaptation to the human innate immune system.


Asunto(s)
Chlamydia trachomatis/inmunología , Chlamydia trachomatis/fisiología , Células Epiteliales/inmunología , Interacciones Huésped-Patógeno , Interferón gamma/inmunología , Células A549 , Animales , Chlamydia muridarum/inmunología , Chlamydia muridarum/fisiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Proteínas de Unión al GTP/metabolismo , Células HeLa , Humanos , Inmunidad Innata , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/microbiología , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Ubiquitinación , Vacuolas/microbiología
4.
Proc Natl Acad Sci U S A ; 112(41): E5628-37, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26417105

RESUMEN

Many microbes create and maintain pathogen-containing vacuoles (PVs) as an intracellular niche permissive for microbial growth and survival. The destruction of PVs by IFNγ-inducible guanylate binding protein (GBP) and immunity-related GTPase (IRG) host proteins is central to a successful immune response directed against numerous PV-resident pathogens. However, the mechanism by which IRGs and GBPs cooperatively detect and destroy PVs is unclear. We find that host cell priming with IFNγ prompts IRG-dependent association of Toxoplasma- and Chlamydia-containing vacuoles with ubiquitin through regulated translocation of the E3 ubiquitin ligase tumor necrosis factor (TNF) receptor associated factor 6 (TRAF6). This initial ubiquitin labeling elicits p62-mediated escort and deposition of GBPs to PVs, thereby conferring cell-autonomous immunity. Hypervirulent strains of Toxoplasma gondii evade this process via specific rhoptry protein kinases that inhibit IRG function, resulting in blockage of downstream PV ubiquitination and GBP delivery. Our results define a ubiquitin-centered mechanism by which host cells deliver GBPs to PVs and explain how hypervirulent parasites evade GBP-mediated immunity.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , Proteínas de Unión al GTP/inmunología , Evasión Inmune , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Ubiquitina/inmunología , Vacuolas/inmunología , Animales , Proteínas de Unión al GTP/genética , Inmunidad Innata , Ratones , Ratones Noqueados , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Toxoplasmosis/genética , Toxoplasmosis/patología , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Vacuolas/metabolismo , Vacuolas/microbiología
5.
Infect Immun ; 83(12): 4740-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416908

RESUMEN

Interferon (IFN)-inducible guanylate binding proteins (GBPs) mediate cell-autonomous host resistance to bacterial pathogens and promote inflammasome activation. The prevailing model postulates that these two GBP-controlled activities are directly linked through GBP-dependent vacuolar lysis. It was proposed that the rupture of pathogen-containing vacuoles (PVs) by GBPs destroyed the microbial refuge and simultaneously contaminated the host cell cytosol with microbial activators of inflammasomes. Here, we demonstrate that GBP-mediated host resistance and GBP-mediated inflammatory responses can be uncoupled. We show that PVs formed by the rodent pathogen Chlamydia muridarum, so-called inclusions, remain free of GBPs and that C. muridarum is impervious to GBP-mediated restrictions on bacterial growth. Although GBPs neither bind to C. muridarum inclusions nor restrict C. muridarum growth, we find that GBPs promote inflammasome activation in C. muridarum-infected macrophages. We demonstrate that C. muridarum infections induce GBP-dependent pyroptosis through both caspase-11-dependent noncanonical and caspase-1-dependent canonical inflammasomes. Among canonical inflammasomes, we find that C. muridarum and the human pathogen Chlamydia trachomatis activate not only NLRP3 but also AIM2. Our data show that GBPs support fast-kinetics processing and secretion of interleukin-1ß (IL-1ß) and IL-18 by the NLRP3 inflammasome but are dispensable for the secretion of the same cytokines at later times postinfection. Because IFN-γ fails to induce IL-1ß transcription, GBP-dependent fast-kinetics inflammasome activation can drive the preferential processing of constitutively expressed IL-18 in IFN-γ-primed macrophages in the absence of prior Toll-like receptor stimulation. Together, our results reveal that GBPs control the kinetics of inflammasome activation and thereby shape macrophage responses to Chlamydia infections.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia muridarum/inmunología , Proteínas de Unión al GTP/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia muridarum/genética , Chlamydia muridarum/patogenicidad , Chlamydia trachomatis/genética , Chlamydia trachomatis/inmunología , Chlamydia trachomatis/patogenicidad , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Fibroblastos/inmunología , Fibroblastos/microbiología , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Cuerpos de Inclusión/inmunología , Cuerpos de Inclusión/microbiología , Inflamasomas/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Cultivo Primario de Células , Transducción de Señal , Vacuolas/inmunología , Vacuolas/microbiología
6.
Commun Integr Biol ; 8(6): e1115163, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27066178

RESUMEN

Many intracellular bacterial and protozoan pathogens reside within host cell vacuoles customized by the microbial invaders to fit their needs. Within such pathogen-containing vacuoles (PVs) microbes procure nutrients and simultaneously hide from cytosolic host defense systems. Among the many PV-resident human pathogens are the bacterium Chlamydia trachomatis and the protozoan Toxoplasma gondii. Immune responses directed against their PVs are poorly characterized. We reported that activation of host cells with IFNγ triggers the attachment of polyubiquitin chains to Toxoplasma- and Chlamydia-containing vacuoles and thereby marks PVs for destruction. In murine cells PV ubiquitination is dependent on IFNγ-inducible Immunity Related GTPases (IRGs). Human cells also decorate PVs with ubiquitin upon IFNγ priming; however, the molecular machinery promoting PV ubiquitination in human cells remains unknown and is likely to be distinct from the IRG-dependent pathway we described in murine cells. Thus, IFNγ-inducible PV ubiquitination constitutes a critical event in cell-autonomous immunity to C. trachomatis and T. gondii in mice and humans, but the molecular machinery underlying PV ubiquitination is expected to be multifaceted and possibly host species-specific.

7.
Proc Natl Acad Sci U S A ; 111(16): 6046-51, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24715728

RESUMEN

IFN receptor signaling induces cell-autonomous immunity to infections with intracellular bacterial pathogens. Here, we demonstrate that IFN-inducible guanylate binding protein (Gbp) proteins stimulate caspase-11-dependent, cell-autonomous immunity in response to cytoplasmic LPS. Caspase-11-dependent pyroptosis is triggered in IFN-activated macrophages infected with the Gram-negative bacterial pathogen Legionella pneumophila. The rapid induction of pyroptosis in IFN-activated macrophages required a cluster of IFN-inducible Gbp proteins encoded on mouse chromosome 3 (Gbp(chr3)). Induction of pyroptosis in naive macrophages by infections with the cytosol-invading ΔsdhA L. pneumophila mutant was similarly dependent on Gbp(chr3), suggesting that these Gbp proteins play a role in the detection of bacteria accessing the cytosol. Cytoplasmic LPS derived from Salmonella ssp. or Escherichia coli has recently been shown to trigger caspase-11 activation and pyroptosis, but the cytoplasmic sensor for LPS and components of the caspase-11 inflammasome are not yet defined. We found that the induction of caspase-11-dependent pyroptosis by cytoplasmic L. pneumophila-derived LPS required Gbp(chr3) proteins. Similarly, pyroptosis induced by cytoplasmic LPS isolated from Salmonella was diminished in Gbp(chr3)-deficient macrophages. These data suggest a role for Gbp(chr3) proteins in the detection of cytoplasmic LPS and the activation of the noncanonical inflammasome.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al GTP/metabolismo , Lipopolisacáridos/farmacología , Animales , Caspasas Iniciadoras , Citoplasma/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Interferón gamma/farmacología , Legionella pneumophila/efectos de los fármacos , Legionella pneumophila/crecimiento & desarrollo , Legionella pneumophila/fisiología , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación/genética , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/fisiología
8.
PLoS One ; 9(1): e86684, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466199

RESUMEN

Cell-autonomous immunity to the bacterial pathogen Chlamydia trachomatis and the protozoan pathogen Toxoplasma gondii is controlled by two families of Interferon (IFN)-inducible GTPases: Immunity Related GTPases (IRGs) and Guanylate binding proteins (Gbps). Members of these two GTPase families associate with pathogen-containing vacuoles (PVs) and solicit antimicrobial resistance pathways specifically to the intracellular site of infection. The proper delivery of IRG and Gbp proteins to PVs requires the autophagy factor Atg5. Atg5 is part of a protein complex that facilitates the transfer of the ubiquitin-like protein Atg8 from the E2-like conjugation enzyme Atg3 to the lipid phosphatidylethanolamine. Here, we show that Atg3 expression, similar to Atg5 expression, is required for IRG and Gbp proteins to dock to PVs. We further demonstrate that expression of a dominant-active, GTP-locked IRG protein variant rescues the PV targeting defect of Atg3- and Atg5-deficient cells, suggesting a possible role for Atg proteins in the activation of IRG proteins. Lastly, we show that IFN-induced cell-autonomous resistance to C. trachomatis infections in mouse cells depends not only on Atg5 and IRG proteins, as previously demonstrated, but also requires the expression of Atg3 and Gbp proteins. These findings provide a foundation for a better understanding of IRG- and Gbp-dependent cell-autonomous resistance and its regulation by Atg proteins.


Asunto(s)
Chlamydia trachomatis/metabolismo , Resistencia a la Enfermedad , Proteínas de Unión al GTP/metabolismo , Inmunidad , Toxoplasma/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Vacuolas/metabolismo , Animales , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/metabolismo , Infecciones por Chlamydia/patología , Chlamydia trachomatis/efectos de los fármacos , Cromosomas de los Mamíferos/metabolismo , Resistencia a la Enfermedad/efectos de los fármacos , Guanosina Trifosfato/metabolismo , Inmunidad/efectos de los fármacos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Interferón gamma/farmacología , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mutantes/metabolismo , Unión Proteica/efectos de los fármacos , Toxoplasma/efectos de los fármacos , Toxoplasmosis/inmunología , Toxoplasmosis/metabolismo , Toxoplasmosis/patología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Vacuolas/efectos de los fármacos
9.
PLoS Pathog ; 9(6): e1003414, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785284

RESUMEN

Interferon-inducible GTPases of the Immunity Related GTPase (IRG) and Guanylate Binding Protein (GBP) families provide resistance to intracellular pathogenic microbes. IRGs and GBPs stably associate with pathogen-containing vacuoles (PVs) and elicit immune pathways directed at the targeted vacuoles. Targeting of Interferon-inducible GTPases to PVs requires the formation of higher-order protein oligomers, a process negatively regulated by a subclass of IRG proteins called IRGMs. We found that the paralogous IRGM proteins Irgm1 and Irgm3 fail to robustly associate with "non-self" PVs containing either the bacterial pathogen Chlamydia trachomatis or the protozoan pathogen Toxoplasma gondii. Instead, Irgm1 and Irgm3 reside on "self" organelles including lipid droplets (LDs). Whereas IRGM-positive LDs are guarded against the stable association with other IRGs and GBPs, we demonstrate that IRGM-stripped LDs become high affinity binding substrates for IRG and GBP proteins. These data reveal that intracellular immune recognition of organelle-like structures by IRG and GBP proteins is partly dictated by the missing of "self" IRGM proteins from these structures.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , GTP Fosfohidrolasas/inmunología , Proteínas de Unión al GTP/inmunología , Inmunidad Innata , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Vacuolas/inmunología , Animales , Línea Celular , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Ratones , Ratones Noqueados , Toxoplasma/metabolismo , Toxoplasmosis/genética , Toxoplasmosis/metabolismo , Vacuolas/genética , Vacuolas/metabolismo , Vacuolas/microbiología , Vacuolas/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...