Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 83(21): 3885-3903.e5, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37832545

RESUMEN

The translocation of stimulator of interferon genes (STING) from the endoplasmic reticulum (ER) to the ER-Golgi intermediate compartment (ERGIC) enables its activation. However, the mechanism underlying the regulation of STING exit from the ER remains elusive. Here, we found that STING induces the activation of transforming growth factor beta-activated kinase 1 (TAK1) prior to STING trafficking in a TAK1 binding protein 1 (TAB1)-dependent manner. Intriguingly, activated TAK1 directly mediates STING phosphorylation on serine 355, which facilitates its interaction with STING ER exit protein (STEEP) and thereby promotes its oligomerization and translocation to the ERGIC for subsequent activation. Importantly, activation of TAK1 by monophosphoryl lipid A, a TLR4 agonist, boosts cGAMP-induced antitumor immunity dependent on STING phosphorylation in a mouse allograft tumor model. Taken together, TAK1 was identified as a checkpoint for STING activation by promoting its trafficking, providing a basis for combinatory tumor immunotherapy and intervention in STING-related diseases.


Asunto(s)
Neoplasias , Animales , Ratones , Fosforilación
2.
Mol Cell ; 82(11): 2032-2049.e7, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35460603

RESUMEN

Virus infection modulates both host immunity and host genomic stability. Poly(ADP-ribose) polymerase 1 (PARP1) is a key nuclear sensor of DNA damage, which maintains genomic integrity, and the successful application of PARP1 inhibitors for clinical anti-cancer therapy has lasted for decades. However, precisely how PARP1 gains access to cytoplasm and regulates antiviral immunity remains unknown. Here, we report that DNA virus induces a reactive nitrogen species (RNS)-dependent DNA damage and activates DNA-dependent protein kinase (DNA-PK). Activated DNA-PK phosphorylates PARP1 on Thr594, thus facilitating the cytoplasmic translocation of PARP1 to inhibit the antiviral immunity both in vitro and in vivo. Mechanistically, cytoplasmic PARP1 interacts with and directly PARylates cyclic GMP-AMP synthase (cGAS) on Asp191 to inhibit its DNA-binding ability. Together, our findings uncover an essential role of PARP1 in linking virus-induced genome instability with inhibition of host immunity, which is of relevance to cancer, autoinflammation, and other diseases.


Asunto(s)
Antivirales , Nucleotidiltransferasas , Antivirales/farmacología , Citoplasma/genética , Citoplasma/metabolismo , ADN , Daño del ADN , Inestabilidad Genómica , Humanos , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo
3.
Nat Immunol ; 17(4): 397-405, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26928339

RESUMEN

The signaling adaptor TRAF3 is a highly versatile regulator of both innate immunity and adaptive immunity, but how its phosphorylation is regulated is still unknown. Here we report that deficiency in or inhibition of the conserved serine-threonine kinase CK1ɛ suppressed the production of type I interferon in response to viral infection. CK1ɛ interacted with and phosphorylated TRAF3 at Ser349, which thereby promoted the Lys63 (K63)-linked ubiquitination of TRAF3 and subsequent recruitment of the kinase TBK1 to TRAF3. Consequently, CK1ɛ-deficient mice were more susceptible to viral infection. Our findings establish CK1ɛ as a regulator of antiviral innate immune responses and indicate a novel mechanism of immunoregulation that involves CK1ɛ-mediated phosphorylation of TRAF3.


Asunto(s)
Caseína Cinasa 1 épsilon/inmunología , Inmunidad Innata/inmunología , Interferón beta/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Animales , Caseína Cinasa 1 épsilon/antagonistas & inhibidores , Caseína Cinasa 1 épsilon/genética , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Células HeLa , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Humanos , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Interferón beta/biosíntesis , Espectrometría de Masas , Ratones , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones por Rhabdoviridae/inmunología , Factor 3 Asociado a Receptor de TNF/genética , Ubiquitinación , Vesiculovirus/inmunología , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología
4.
Nature ; 577(7792): 682-688, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31942069

RESUMEN

Mycobacterium tuberculosis is an intracellular pathogen that uses several strategies to interfere with the signalling functions of host immune molecules. Many other bacterial pathogens exploit the host ubiquitination system to promote pathogenesis1,2, but whether this same system modulates the ubiquitination of M. tuberculosis proteins is unknown. Here we report that the host E3 ubiquitin ligase ANAPC2-a core subunit of the anaphase-promoting complex/cyclosome-interacts with the mycobacterial protein Rv0222 and promotes the attachment of lysine-11-linked ubiquitin chains to lysine 76 of Rv0222 in order to suppress the expression of proinflammatory cytokines. Inhibition of ANAPC2 by specific short hairpin RNA abolishes the inhibitory effect of Rv0222 on proinflammatory responses. Moreover, mutation of the ubiquitination site on Rv0222 impairs the inhibition of proinflammatory cytokines by Rv0222 and reduces virulence during infection in mice. Mechanistically, lysine-11-linked ubiquitination of Rv0222 by ANAPC2 facilitates the recruitment of the protein tyrosine phosphatase SHP1 to the adaptor protein TRAF6, preventing the lysine-63-linked ubiquitination and activation of TRAF6. Our findings identify a previously unrecognized mechanism that M. tuberculosis uses to suppress host immunity, and provide insights relevant to the development of effective immunomodulators that target M. tuberculosis.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Ubiquitinación , Ciclosoma-Complejo Promotor de la Anafase/química , Animales , Subunidad Apc2 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Lisina/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción AP-1/metabolismo , Tuberculosis/microbiología , Virulencia/inmunología
5.
Nat Immunol ; 13(11): 1063-71, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23001144

RESUMEN

The protein Tir (translocated intimin receptor) in enteric bacteria shares sequence similarity with the host cellular immunoreceptor tyrosine-based inhibition motifs (ITIMs). Despite the importance of Tir in pedestal formation, relatively little is known about the role of Tir and its ITIMs in the regulation of the host immune response. Here we demonstrate that Tir from enteropathogenic Escherichia coli (EPEC) interacted with the host cellular tyrosine phosphatase SHP-1 in an ITIM phosphorylation-dependent manner. The association of Tir with SHP-1 facilitated the recruitment of SHP-1 to the adaptor TRAF6 and inhibited the ubiquitination of TRAF6. Moreover, the ITIMs of Tir suppressed EPEC-stimulated expression of proinflammatory cytokines and inhibited intestinal immunity to infection with Citrobacter rodentium. Our findings identify a previously unknown mechanism by which bacterial ITIM-containing proteins can inhibit innate immune responses.


Asunto(s)
Infecciones por Enterobacteriaceae/inmunología , Proteínas de Escherichia coli/inmunología , Motivo de Inhibición del Inmunorreceptor Basado en Tirosina/inmunología , Intestinos/inmunología , Receptores de Superficie Celular/inmunología , Transducción de Señal/inmunología , Animales , Células Cultivadas , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enteropatógena/inmunología , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/genética , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata , Inmunidad Mucosa , Motivo de Inhibición del Inmunorreceptor Basado en Tirosina/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Receptores de Superficie Celular/genética , Transducción de Señal/genética , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Ubiquitina/genética , Ubiquitina/inmunología , Ubiquitinación
6.
Nature ; 563(7729): 131-136, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30356214

RESUMEN

Accurate repair of DNA double-stranded breaks by homologous recombination preserves genome integrity and inhibits tumorigenesis. Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that activates innate immunity by initiating the STING-IRF3-type I IFN signalling cascade1,2. Recognition of ruptured micronuclei by cGAS links genome instability to the innate immune response3,4, but the potential involvement of cGAS in DNA repair remains unknown. Here we demonstrate that cGAS inhibits homologous recombination in mouse and human models. DNA damage induces nuclear translocation of cGAS in a manner that is dependent on importin-α, and the phosphorylation of cGAS at tyrosine 215-mediated by B-lymphoid tyrosine kinase-facilitates the cytosolic retention of cGAS. In the nucleus, cGAS is recruited to double-stranded breaks and interacts with PARP1 via poly(ADP-ribose). The cGAS-PARP1 interaction impedes the formation of the PARP1-Timeless complex, and thereby suppresses homologous recombination. We show that knockdown of cGAS suppresses DNA damage and inhibits tumour growth both in vitro and in vivo. We conclude that nuclear cGAS suppresses homologous-recombination-mediated repair and promotes tumour growth, and that cGAS therefore represents a potential target for cancer prevention and therapy.


Asunto(s)
Núcleo Celular/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias/metabolismo , Neoplasias/patología , Nucleotidiltransferasas/metabolismo , Reparación del ADN por Recombinación , Transporte Activo de Núcleo Celular , Adulto , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Núcleo Celular/enzimología , Roturas del ADN de Doble Cadena , Daño del ADN , Femenino , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Nucleotidiltransferasas/deficiencia , Fosforilación , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Unión Proteica/efectos de los fármacos , Reparación del ADN por Recombinación/genética , Familia-src Quinasas/metabolismo
7.
EMBO Rep ; 22(7): e51678, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33987949

RESUMEN

Mycobacterial arabinogalactan (AG) is an essential cell wall component of mycobacteria and a frequent structural and bio-synthetical target for anti-tuberculosis (TB) drug development. Here, we report that mycobacterial AG is recognized by galectin-9 and exacerbates mycobacterial infection. Administration of AG-specific aptamers inhibits cellular infiltration caused by Mycobacterium tuberculosis (Mtb) or Mycobacterium bovis BCG, and moderately increases survival of Mtb-infected mice or Mycobacterium marinum-infected zebrafish. AG interacts with carbohydrate recognition domain (CRD) 2 of galectin-9 with high affinity, and galectin-9 associates with transforming growth factor ß-activated kinase 1 (TAK1) via CRD2 to trigger subsequent activation of extracellular signal-regulated kinase (ERK) as well as induction of the expression of matrix metalloproteinases (MMPs). Moreover, deletion of galectin-9 or inhibition of MMPs blocks AG-induced pathological impairments in the lung, and the AG-galectin-9 axis aggravates the process of Mtb infection in mice. These results demonstrate that AG is an important virulence factor of mycobacteria and galectin-9 is a novel receptor for Mtb and other mycobacteria, paving the way for the development of novel effective TB immune modulators.


Asunto(s)
Mycobacterium tuberculosis , Pez Cebra , Animales , Galactanos , Galectinas/genética , Ratones
8.
EMBO Rep ; 20(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30872316

RESUMEN

Cyclic dinucleotides (CDNs) are important second messenger molecules in prokaryotes and eukaryotes. Within host cells, cytosolic CDNs are detected by STING and alert the host by activating innate immunity characterized by type I interferon (IFN) responses. Extracellular bacteria and dying cells can release CDNs, but sensing of extracellular CDNs (eCDNs) by mammalian cells remains elusive. Here, we report that endocytosis facilitates internalization of eCDNs. The DNA sensor cGAS facilitates sensing of endocytosed CDNs, their perinuclear accumulation, and subsequent STING-dependent release of type I IFN Internalized CDNs bind cGAS directly, leading to its dimerization, and the formation of a cGAS/STING complex, which may activate downstream signaling. Thus, eCDNs comprise microbe- and danger-associated molecular patterns that contribute to host-microbe crosstalk during health and disease.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , Animales , Línea Celular , Endocitosis/genética , Endocitosis/inmunología , Espacio Extracelular , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón Tipo I/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Modelos Moleculares , Nucleótidos Cíclicos/química , Nucleotidiltransferasas/química , Nucleotidiltransferasas/genética , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Sistemas de Mensajero Secundario , Transducción de Señal , Relación Estructura-Actividad
9.
J Cell Mol Med ; 24(21): 12716-12725, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32977368

RESUMEN

The role of corticosteroids in acute lung injury (ALI) remains uncertain. This study aims to determine the underlying mechanisms of corticosteroid treatment for lipopolysaccharide (LPS)-induced inflammation and ALI. We used corticosteroid treatment for LPS-induced murine ALI model to investigate the effect of corticosteroid on ALI in vivo. Moreover, LPS-stimulated macrophages were used to explore the specific anti-inflammatory effects of corticosteroids on NLRP3-inflammasome in vitro. We found corticosteroids attenuated LPS-induced ALI, which manifested in reduction of the alveolar structure destruction, the infiltration of neutrophils and the inflammatory cytokines release of interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) in Lung. In vitro, when NLRP3-inflammasome was knocked out, inflammatory response of caspase-1 activation and IL-1ß secretion was obviously declined. Further exploration, our results showed that when corticosteroid preprocessed macrophages before LPS primed, it obviously inhibited the activation of caspase-1 and the maturation of IL-1ß, which depended on inhibiting the nuclear factor-κB (NF-κB) signal pathway activation. However, when corticosteroids intervened the LPS-primed macrophages, it also negatively regulated NLRP3-inflammasome activation through suppressing mitochondrial reactive oxygen species (mtROS) production. Our results revealed that corticosteroids played a protection role in LPS-induced inflammation and ALI by suppressing both NF-κB signal pathway and mtROS-dependent NLRP3 inflammasome activation.


Asunto(s)
Corticoesteroides/uso terapéutico , Inflamasomas/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Lesión Pulmonar Aguda , Corticoesteroides/farmacología , Animales , Caspasa 1/metabolismo , Dexametasona/farmacología , Dexametasona/uso terapéutico , Activación Enzimática/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Interleucina-18/metabolismo , Lipopolisacáridos , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
10.
Nat Immunol ; 9(8): 898-907, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18604210

RESUMEN

The inhibitory signaling of natural killer (NK) cells is crucial in the regulation of innate immune responses. Here we show that the association of KIR2DL1, an inhibitory receptor of NK cells, with beta-arrestin 2 mediated recruitment of the tyrosine phosphatases SHP-1 and SHP-2 to KIR2DL1 and facilitated 'downstream' inhibitory signaling. Consequently, the cytotoxicity of NK cells was higher in beta-arrestin 2-deficient mice but was inhibited in beta-arrestin 2-transgenic mice. Moreover, beta-arrestin 2-deficient mice were less susceptible than wild-type mice to mouse cytomegalovirus infection, an effect that was abolished by depletion of NK cells. Our findings identify a previously unknown mechanism by which the inhibitory signaling in NK cells is regulated.


Asunto(s)
Arrestinas/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología , Animales , Células Cultivadas , Péptidos y Proteínas de Señalización Intracelular , Ratones , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Arrestina beta 2 , beta-Arrestinas
11.
Arch Biochem Biophys ; 694: 108612, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33007281

RESUMEN

Apoptotic and inflammatory pathways play important roles in Mycobacterium tuberculosis-infected macrophages. Sirt1 is a member of the deacetylase family that is known to promote apoptosis resistance in mammalian cells and was recently reported to regulate mycobacterial immunopathogenesis via inflammatory responses. However, the apoptotic role of Sirt1 in the process of M. tuberculosis infection remains unclear. With the help of mouse peritoneal macrophage samples, we have shown that resveratrol, a Sirt1 activator, inhibited M. tuberculosis-induced apoptosis in peritoneal macrophages. Further, we found that Sirt1 activation prompted M. tuberculosis induced GSK3ß phosphorylation. Further investigation into the possible mechanisms of action showed that Sirt1 directly interacted with GSK3ß and enhanced GSK3ß phosphorylation by promoting its deacetylation. Sirt1 activation inhibited M. tuberculosis growth. Thus, it seemed that Sirt1 acted as a novel regulator of apoptosis signaling in M. tuberculosis infection via its direct effects on GSK3ß. Sirt1 may therefore be a new candidate for the prevention and treatment of tuberculosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Macrófagos/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Sirtuina 1/metabolismo , Animales , Activadores de Enzimas/farmacología , Femenino , Glucógeno Sintasa Quinasa 3 beta/química , Células HEK293 , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Mycobacterium tuberculosis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Resveratrol/farmacología , Transducción de Señal/efectos de los fármacos
12.
BMC Biol ; 17(1): 7, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683096

RESUMEN

BACKGROUND: The emergence of drug-resistant strains of Mycobacterium tuberculosis (Mtb), especially those that are multidrug resistant poses a serious threat to global tuberculosis control. However, the mechanism underlying the occurrence of drug resistance against more than one drug is poorly understood. Given that the Beijing/W strains are associated with outbreaks and multidrug resistance, they may harbor a genetic advantage and provide useful insight into the disease. One marker found in all Beijing/W Mtb strains is a deletion of RD105 region that results in a gene fusion, Rv0071/74, with a variable number (3-9 m) of VDP (V: Val, D: Asp; P: Pro) repeats (coded by gtggacccg repeat sequences) at the N-terminal. Here, we report that this variable number of VDP repeats in Rv0071/74 regulates the development of multidrug resistance. RESULTS: We collected and analyzed 1255 Beijing/W clinical strains. The results showed that the number of VDP repeats in Rv0071/74 was related to the development of multidrug resistance, and the deletion of Rv0071/74-9 m from Beijing/W clinical strain restored drug susceptibility. Rv0071/74-9 m also increased resistance to multiple drugs when transferred to different mycobacterial strains. Cell-free assays indicate that the domain carrying 4-9 VDP repeats (4-9 m) showed a variable binding affinity with peptidoglycan and Rv0071/74 cleaves peptidoglycan. Furthermore, Rv0071/74-9 m increased cell wall thickness and reduced the intracellular concentration of antibiotics. CONCLUSIONS: These findings not only identify Rv0071/74 with VDP repeats as a newly identified multidrug resistance gene but also provide a new model for the development of multiple drug resistance.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple/genética , Mycobacterium tuberculosis/genética , Eliminación de Secuencia , Genotipo , Mycobacterium tuberculosis/efectos de los fármacos
13.
Arch Biochem Biophys ; 667: 49-58, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31029687

RESUMEN

Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) has an alarming mortality rate, and the fast-growing drug resistance of Mtb necessitates the discovery and application of new therapeutic agents. Although Sirt1 is a key regulator in metabolic, cardiovascular and other age-related diseases, its role in the regulation of antibacterial immunity is unclear. Here, we have reported that Sirt1 expression was decreased, and the decreased Sirt1 level was correlated with increased pro-flammatory cytokines and TAK1 phosphorylation in active TB patients. Our mechanistic experiments showed that Sirt1 direclty interacted with TAK1 and suppressed its activation. Further, Sirt1+/- macrophages infected with Mtb exhibited enhanced activation of TAK1, MAPKs and NF-κB, as well as simultaneously elevated expression of pro-inflammatory cytokines. Moreover, Sirt1+/- mice exhibited overt inflammation, as indicated by the significant abundance of pro-inflammatory cytokines, and were more susceptible to Mtb infection than wild-type mice. Overall, the findings indicate that inhibition of Sirt1 expression by Mtb infection enhances TAK1 activation, and this in turn enhances the secretion of IL-6 and TNF-α via activation of the p65/p38/JNK/ERK signaling pathways. Treatment with resveratrol, which is known that one of its multiple effects is Sirt1 activation, in Mtb-infected macrophages inhibited the activation of TAK1, MAPK and NF-κB pathways, and the pro-inflammatory cytokine levels. Consitently, mice treated with resveratrol were more resistant to Mtb infection. The potent therapeutic effects of resveratrol against Mtb infection indicate that Sirt1 could be a novel therapeutic target for the treatment of TB.


Asunto(s)
Inmunidad Innata , Quinasas Quinasa Quinasa PAM/inmunología , Mycobacterium tuberculosis/inmunología , Sirtuina 1/inmunología , Adulto , Animales , Citocinas/biosíntesis , Activación Enzimática/inmunología , Femenino , Células HEK293 , Humanos , Técnicas In Vitro , Mediadores de Inflamación/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/microbiología , Masculino , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Resveratrol/farmacología , Transducción de Señal , Sirtuina 1/deficiencia , Sirtuina 1/genética , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/enzimología , Tuberculosis Pulmonar/inmunología
14.
J Immunol ; 198(12): 4772-4780, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28507027

RESUMEN

Tuberculosis caused by Mycobacterium tuberculosis continues to pose a serious global health threat. The attenuated Mycobacterium bovis bacillus Calmette-Guérin, as the only licensed vaccine, has limited protective efficacy against TB. The development of more effective antituberculosis vaccines is urgent and demands for further identification and understanding of M. tuberculosis Ags. MPT83 (Rv2873), a secreted mycobacterial lipoprotein, has been applied into subunit vaccine development and shown protective effects against M. tuberculosis infection in animals; however, the understanding of the underlying mechanism is limited. In present study, we systematically studied the effect of MPT83 on macrophage apoptosis by constructing Mycobacterium smegmatis strain overexpressing MPT83 (MS_MPT83) and purifying rMPT83 protein. We found that MPT83 induced apoptosis in both human and mouse macrophages. MPT83 induced cyclooxygenase-2 (COX-2) expression at both the transcriptional and protein levels in macrophages, whereas silencing or inhibiting COX-2 blocked rMPT83-induced apoptosis or the enhanced apoptotic response to MS_MPT83 in comparison with M. smegmatis transfected with pMV261 vector (MS_Vec), indicating that COX-2 is required for MPT83-induced apoptosis. Additionally, tlr2 deficiency led to significant reduction of COX-2 expression, accompanied by less apoptosis in macrophages stimulated with rMPT83 or infected with MS_MPT83. Moreover, the activation of p38 accounted for MPT83-induced COX-2 expression. Finally, lower bacteria burdens in the lungs and spleens and enhanced survival were observed in mice i.v. infected with MS_MPT83 compared with MS_Vec. Taken together, our results established a proapoptotic effect of MPT83 and identified the TLR2/p38/COX-2 axis in MPT83-induced macrophage apoptosis.


Asunto(s)
Antígenos Bacterianos/fisiología , Apoptosis , Proteínas Bacterianas/fisiología , Ciclooxigenasa 2/metabolismo , Macrófagos/microbiología , Macrófagos/fisiología , Proteínas de la Membrana/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Ciclooxigenasa 2/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/aislamiento & purificación , Ratones , Mycobacterium smegmatis/inmunología , Fosforilación , Receptor Toll-Like 2/inmunología
15.
J Infect Dis ; 218(2): 312-323, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29228365

RESUMEN

Tuberculosis, caused by Mycobacterium tuberculosis infection, remains a global threat to human health, but knowledge of the molecular mechanisms underlying the pathogenesis of tuberculosis is still limited. Although Notch4, a member of the Notch receptor family, is involved in the initiation of mammary tumors, its function in M. tuberculosis infection remains unclear. In this study, we found that Notch4-deficient mice were more resistant to M. tuberculosis infection, with a much lower bacterial burden and fewer pathological changes in the lungs. Notch4 inhibited M. tuberculosis-induced production of proinflammatory cytokines by interaction with TAK1 and inhibition of its activation. Furthermore, we found that Notch intracellular domain 4 prevented TRAF6 autoubiquitination and suppressed TRAF6-mediated TAK1 polyubiquitination. Finally, Notch inhibitors made mice more resistant to M. tuberculosis infection. These results suggest that Notch4 is a negative regulator of M. tuberculosis-induced inflammatory response, and treatment with a Notch inhibitor could serve as a new therapeutic strategy for tuberculosis.


Asunto(s)
Regulación de la Expresión Génica , Quinasas Quinasa Quinasa PAM/metabolismo , Receptor Notch4/metabolismo , Tuberculosis Pulmonar/patología , Animales , Carga Bacteriana , Citocinas/análisis , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Inflamación/patología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Receptor Notch4/deficiencia , Factor 6 Asociado a Receptor de TNF/metabolismo , Tuberculosis Pulmonar/microbiología
16.
Eur J Immunol ; 47(9): 1414-1426, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28744851

RESUMEN

The innate immune system initiates immune responses by pattern-recognition receptors (PRR). Virus-derived nucleic acids are sensed by the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) family and the toll-like receptor (TLR) family as well as the DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS). These receptors activate IRF3/7 and NF-κB signaling pathways to induce the expression of type I interferons (IFNs) and other cytokines firing antiviral responses within the cell. However, to achieve a favorable outcome for the host, a balanced production of IFNs and activation of antiviral responses is required. Post-translational modifications (PTMs), such as the covalent linkage of functional groups to amino acid chains, are crucial for this immune homeostasis in antiviral responses. Canonical PTMs including phosphorylation and ubiquitination have been extensively studied and other PTMs such as methylation, acetylation, SUMOylation, ADP-ribosylation and glutamylation are being increasingly implicated in antiviral innate immunity. Here we summarize our recent understanding of the most important PTMs regulating the antiviral innate immune response, and their role in virus-related immune pathogenesis.


Asunto(s)
ADN Viral/inmunología , Inmunidad Innata , Procesamiento Proteico-Postraduccional , ARN Viral/inmunología , Virosis/inmunología , Animales , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , FN-kappa B/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
17.
Infect Immun ; 85(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28652310

RESUMEN

Exoenzyme Y (ExoY) is a type III secretion system effector found in 90% of the Pseudomonas aeruginosa isolates. Although it is known that ExoY is a soluble nucleotidyl cyclase that increases the cytoplasmic levels of nucleoside 3',5'-cyclic monophosphates (cNMPs) to mediate endothelial Tau phosphorylation and permeability, its functional role in the innate immune response is still poorly understood. Transforming growth factor ß-activated kinase 1 (TAK1) is critical for mediating Toll-like receptor (TLR) signaling and subsequent activation of NF-κB and AP-1, which are transcriptional activators of innate immunity. Here, we report that ExoY inhibits proinflammatory cytokine production through suppressing the activation of TAK1 as well as downstream NF-κB and mitogen-activated protein (MAP) kinases. Mice infected with ExoY-deficient P. aeruginosa had higher levels of tumor necrosis factor (TNF) and interleukin-6 (IL-6), more neutrophil recruitment, and a lower bacterial load in lung tissue than mice infected with wild-type P. aeruginosa Taken together, our findings identify a previously unknown mechanism by which P. aeruginosa ExoY inhibits the host innate immune response.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glucosiltransferasas/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Inmunidad Innata , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Liasas de Fósforo-Oxígeno/metabolismo , Pseudomonas aeruginosa/patogenicidad , Animales , Proteínas Bacterianas/genética , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Glucosiltransferasas/genética , Humanos , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Liasas de Fósforo-Oxígeno/genética , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/enzimología , Factor de Transcripción AP-1/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L677-L686, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28684545

RESUMEN

Macrolides antibiotics have been effectively used in many chronic diseases, especially with Pseudomonas aeruginosa (P. aeruginosa) infection. The mechanisms underlying the therapeutic effects of macrolides in these diseases remain poorly understood. We established a mouse model of chronic lung infection using P. aeruginosa agar-beads, with azithromycin treatment or placebo. Lung injury, bacterial clearance, and inflammasome-related proteins were measured. In vitro, the inflammasomes activation induced by flagellin or ATP were assessed in LPS-primed macrophages with or without macrolides treatment. Plasma IL-18 levels were determined from patients who were diagnosed with bronchiectasis isolated with or without P. aeruginosa and treated with azithromycin for 3-5 days. Azithromycin treatment enhanced bacterial clearance and attenuated lung injury in mice chronically infected with P. aeruginosa, which resulted from the inhibition of caspase-1-dependent IL-1ß and IL-18 secretion. In vitro, azithromycin and erythromycin inhibited NLRC4 and NLRP3 inflammasomes activation. Plasma IL-18 levels were higher in bronchiectasis patients with P. aeruginosa isolation compared with healthy controls. Azithromycin administration markedly decreased IL-18 secretion in bronchiectasis patients. The results of this study reveal that azithromycin and erythromycin exert a novel anti-inflammatory effect by attenuating inflammasomes activation, which suggests potential treatment options for inflammasome-related diseases.


Asunto(s)
Bronquiectasia/tratamiento farmacológico , Inflamasomas/antagonistas & inhibidores , Macrólidos/farmacología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Antibacterianos/farmacología , Azitromicina/farmacología , Bronquiectasia/microbiología , Células Cultivadas , Humanos , Inflamasomas/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/microbiología
19.
PLoS Pathog ; 11(1): e1004613, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25615690

RESUMEN

Enterovirus 71 (EV71) is the most virulent pathogen among enteroviruses that cause hand, foot and mouth disease in children but rarely in adults. The mechanisms that determine the age-dependent susceptibility remain largely unclear. Here, we found that the paucity of invariant natural killer T (iNKT) cells together with immaturity of the immune system was related to the susceptibility of neonatal mice to EV71 infection. iNKT cells were crucial antiviral effector cells to protect young mice from EV71 infection before their adaptive immune systems were fully mature. EV71 infection led to activation of iNKT cells depending on signaling through TLR3 but not other TLRs. Surprisingly, iNKT cell activation during EV71 infection required TLR3 signaling in macrophages, but not in dendritic cells (DCs). Mechanistically, interleukin (IL)-12 and endogenous CD1d-restricted antigens were both required for full activation of iNKT cells. Furthermore, CD1d-deficiency led to dramatically increased viral loads in central nervous system and more severe disease in EV71-infected mice. Altogether, our results suggest that iNKT cells may be involved in controlling EV71 infection in children when their adaptive immune systems are not fully developed, and also imply that iNKT cells might be an intervention target for treating EV71-infected patients.


Asunto(s)
Enterovirus Humano A/inmunología , Infecciones por Enterovirus/inmunología , Inmunidad Celular , Macrófagos/inmunología , Células T Asesinas Naturales/inmunología , Receptor Toll-Like 3/fisiología , Animales , Células Cultivadas , Infecciones por Enterovirus/genética , Humanos , Inmunidad Celular/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Células T Asesinas Naturales/metabolismo , Transducción de Señal/inmunología
20.
J Invertebr Pathol ; 149: 66-75, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28782510

RESUMEN

The biopesticide used most effectively to control mosquito and blackfly vectors of human diseases worldwide is Bacillus thuringiensis subsp. israelensis. The high efficacy of this bacterium is due to synergistic interactions among four protein entomotoxins assembled individually into a single parasporal body (PB) during sporulation. Cyt1Aa, the primary synergist, is the most abundant toxin, comprising approximately 55% of the PB's mass. The other proteins are Cry11Aa at ∼35%, and Cry4Aa and Cry4Ba, which together account for the remaining ∼10%. The molecular genetic basis for the comparatively large amount of Cyt1Aa synthesized is unknown. Here, in addition to the known strong BtI (σE) and BtII (σK) promoters, we demonstrate a third promoter (BtIII) that has high identity to the σE promoter of Bacillus subtilis, contributes to the large amount of Cyt1Aa synthesized. We also show that a cyt1Aa-BtIII construct was not functional in a σE-deficient strain of B. subtilis. Comparison of transcription levels and protein profiles for recombinant strains containing different combinations of BtI, BtII and BtIII, or each promoter alone, showed that BtIII is active throughout sporulation. We further demonstrate that a stable stem-loop in the 3'-untranslated region (3'-UTR, predicted ΔG=-27.6) contributes to the high level of Cyt1Aa synthesized.


Asunto(s)
Regiones no Traducidas 3' , Regiones no Traducidas 5' , Bacillus thuringiensis/genética , Proteínas Bacterianas/genética , Endotoxinas/genética , Proteínas Hemolisinas/genética , Toxinas de Bacillus thuringiensis , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA