Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 137, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802820

RESUMEN

Hyperglycemia has been shown to modulate the immune response of peripheral immune cells and organs, but the impact of hyperglycemia on neuroinflammation within the brain remains elusive. In the present study, we provide evidences that streptozotocin (STZ)-induced hyperglycemic condition in mice drives a phenotypic switch of brain astrocytes to a proinflammatory state, and increases brain vulnerability to mild peripheral inflammation. In particular, we found that hyperglycemia led to a significant increase in the astrocyte proliferation as determined by flow cytometric and immunohistochemical analyses of mouse brain. The increased astrocyte proliferation by hyperglycemia was reduced by Glut1 inhibitor BAY-876. Transcriptomic analysis of isolated astrocytes from Aldh1l1CreERT2;tdTomato mice revealed that peripheral STZ injection induced astrocyte reprogramming into proliferative, and proinflammatory phenotype. Additionally, STZ-induced hyperglycemic condition significantly enhanced the infiltration of circulating myeloid cells into the brain and the disruption of blood-brain barrier in response to mild lipopolysaccharide (LPS) administration. Systemic hyperglycemia did not alter the intensity and sensitivity of peripheral inflammation in mice to LPS challenge, but increased the inflammatory potential of brain microglia. In line with findings from mouse experiments, a high-glucose environment intensified the LPS-triggered production of proinflammatory molecules in primary astrocyte cultures. Furthermore, hyperglycemic mice exhibited a significant impairment in cognitive function after mild LPS administration compared to normoglycemic mice as determined by novel object recognition and Y-maze tasks. Taken together, these results demonstrate that hyperglycemia directly induces astrocyte reprogramming towards a proliferative and proinflammatory phenotype, which potentiates mild LPS-triggered inflammation within brain parenchymal regions.


Asunto(s)
Astrocitos , Encéfalo , Hiperglucemia , Lipopolisacáridos , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias , Animales , Hiperglucemia/inducido químicamente , Hiperglucemia/patología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Ratones , Lipopolisacáridos/toxicidad , Lipopolisacáridos/farmacología , Encéfalo/patología , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/inducido químicamente , Masculino , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/fisiología , Ratones Transgénicos , Células Cultivadas
2.
Nat Immunol ; 11(5): 385-93, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20351693

RESUMEN

Francisella tularensis, the causative agent of tularemia, infects host macrophages, which triggers production of the proinflammatory cytokines interleukin 1beta (IL-1beta) and IL-18. We elucidate here how host macrophages recognize F. tularensis and elicit this proinflammatory response. Using mice deficient in the DNA-sensing inflammasome component AIM2, we demonstrate here that AIM2 is required for sensing F. tularensis. AIM2-deficient mice were extremely susceptible to F. tularensis infection, with greater mortality and bacterial burden than that of wild-type mice. Caspase-1 activation, IL-1beta secretion and cell death were absent in Aim2(-/-) macrophages in response to F. tularensis infection or the presence of cytoplasmic DNA. Our study identifies AIM2 as a crucial sensor of F. tularensis infection and provides genetic proof of its critical role in host innate immunity to intracellular pathogens.


Asunto(s)
Francisella tularensis/inmunología , Inmunidad Innata , Macrófagos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Tularemia/inmunología , Animales , Señalización del Calcio/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Proteínas de Unión al ADN , Francisella tularensis/patogenicidad , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/inmunología , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-1beta/inmunología , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/inmunología , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Proteínas Nucleares/genética , Multimerización de Proteína , Tularemia/genética , Tularemia/metabolismo
3.
Bioorg Med Chem Lett ; 65: 128693, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35314328

RESUMEN

A series of N-benzyl 5-(4-sulfamoylbenzylidene-2-thioxothiazolidin-4-one analogs, designed as hybrids of CY09 and JC121, were investigated as inhibitors of NLRP3 inflammasome activation. Among them, compounds 34 and 36 were identified as promising NLRP3 inhibitors by measuring the amount of active caspase-1 p20 and IL-1ß produced by NLRP3 inflammasome activation. Further studies indicated that both compounds inhibited NLRP3 inflammasome assembly by reducing the formation of NLRP3 and ASC oligomer specks and selectively inhibited only NLRP3 inflammasome activation and not other inflammasomes such as NLRC4 and AIM2.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Caspasa 1 , Proteínas de Unión al ADN , Interleucina-1beta
4.
FASEB J ; 33(3): 4547-4558, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30592629

RESUMEN

Endoplasmic reticulum (ER)-Golgi vesicle trafficking plays a pivotal role in the conventional secretory pathway of many cytokines; however, the precise release mechanism of a major inflammasome mediator, IL-1ß, is not thought to follow the conventional ER-Golgi route and remains elusive. Here, we found that perturbation of ER-Golgi trafficking by brefeldin A (BFA) treatment attenuated nucleotide-binding oligomerization domain-like receptor family, pyrin-domain-containing 3 (NLRP3) inflammasome activation in mouse bone marrow-derived macrophages (BMDMs). BFA treatment inhibited NLRP3-mediated inflammasome assembly and caspase-1 activation but did not block IL-1ß secretion from BMDMs following BFA administration after NLRP3 inflammasome activation. Consistently, short-hairpin RNA-dependent knockdown of BFA-inhibited guanine nucleotide-exchange protein 1 (BIG1), a molecular target of BFA and an initiator of Golgi-specific vesicle trafficking, abolished NLRP3-dependent apoptosis-associated speck-like protein containing a caspase-recruitment domain oligomerization and caspase-1 activation in BMDMs. Similarly, knockdown of Golgi-specific BFA-resistance guanine nucleotide exchange factor 1, another target of BFA, clearly attenuated NLRP3-mediated caspase-1 activation in BMDMs. Mechanistically, inhibition of BIG1-mediated vesicle trafficking did not impair NLRP3-activating signal 2-promoted events, such as potassium efflux and mitochondrial rearrangement, but caused significant impairment of signal 1-triggered priming steps, including NF-κB-mediated pathways. These data suggest that BFA-targeted vesicle trafficking at the Golgi contributes to activation of the NLRP3 inflammasome signaling.-Hong, S., Hwang, I., Gim, E., Yang, J., Park, S., Yoon, S.-H., Lee, W.-W., Yu, J.-W. Brefeldin A-sensitive ER-Golgi vesicle trafficking contributes to NLRP3-dependent caspase-1 activation.


Asunto(s)
Brefeldino A/farmacología , Caspasa 1/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Inflamasomas/fisiología , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Transporte de Proteínas/efectos de los fármacos , Adenosina Trifosfato/farmacología , Animales , Retículo Endoplásmico/metabolismo , Activación Enzimática/efectos de los fármacos , Aparato de Golgi/metabolismo , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Inflamasomas/efectos de los fármacos , Interleucina-1beta/biosíntesis , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Potasio/metabolismo , Organismos Libres de Patógenos Específicos , Células THP-1
5.
Proc Natl Acad Sci U S A ; 113(6): E762-71, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26811463

RESUMEN

Commensal microbiota are well known to play an important role in antiviral immunity by providing immune inductive signals; however, the consequence of dysbiosis on antiviral immunity remains unclear. We demonstrate that dysbiosis caused by oral antibiotic treatment directly impairs antiviral immunity following viral infection of the vaginal mucosa. Antibiotic-treated mice succumbed to mucosal herpes simplex virus type 2 infection more rapidly than water-fed mice, and also showed delayed viral clearance at the site of infection. However, innate immune responses, including type I IFN and proinflammatory cytokine production at infection sites, as well as induction of virus-specific CD4 and CD8 T-cell responses in draining lymph nodes, were not impaired in antibiotic-treated mice. By screening the factors controlling antiviral immunity, we found that IL-33, an alarmin released in response to tissue damage, was secreted from vaginal epithelium after the depletion of commensal microbiota. This cytokine suppresses local antiviral immunity by blocking the migration of effector T cells to the vaginal tissue, thereby inhibiting the production of IFN-γ, a critical cytokine for antiviral defense, at local infection sites. These findings provide insight into the mechanisms of homeostasis maintained by commensal bacteria, and reveal a deleterious consequence of dysbiosis in antiviral immune defense.


Asunto(s)
Antivirales/inmunología , Disbiosis/complicaciones , Inmunidad Innata , Interleucina-33/metabolismo , Membrana Mucosa/patología , Vagina/inmunología , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Recuento de Colonia Microbiana , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Femenino , Herpes Genital/inmunología , Herpes Genital/patología , Herpes Genital/virología , Herpesvirus Humano 2/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Interferón gamma/biosíntesis , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Membrana Mucosa/inmunología , Membrana Mucosa/virología , Péptido Hidrolasas/metabolismo , Linfocitos T/efectos de los fármacos , Vagina/efectos de los fármacos , Vagina/patología , Vagina/virología
6.
J Biol Chem ; 292(50): 20437-20448, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29051224

RESUMEN

Advanced glycation end products (AGEs) are adducts formed on proteins by glycation with reducing sugars, such as glucose, and tend to form and accumulate under hyperglycemic conditions. AGE accumulation alters protein function and has been implicated in the pathogenesis of many degenerative diseases such as diabetic complications. AGEs have also been shown to promote the production of pro-inflammatory cytokines, but the roles of AGEs in inflammasome signaling have not been explored in detail. Here, we present evidence that AGEs attenuate activation of the NLRP3 inflammasome in bone marrow-derived macrophages (BMDMs) as determined by caspase-1 processing and interleukin-1ß production. AGEs also dampened the assembly of the NLRP3 inflammasome, but did not affect the NLRC4 or AIM2 inflammasome activation. Moreover, our data indicated that AGE treatment inhibited Toll-like receptor (TLR)-dependent production of pro-inflammatory cytokines in BMDMs. This immunosuppressive effect of AGE was not associated with a receptor for AGEs (RAGE)-mediated signaling. Instead, AGE treatment markedly suppressed lipopolysaccharide-induced M1 polarization of macrophages. Furthermore, AGEs significantly dampened innate immune responses including NLRP3 inflammasome activation and type-I interferon production in macrophages upon influenza virus infection. These observations collectively suggest that AGEs could impair host NLRP3 inflammasome-mediated innate immune defenses against RNA virus infection leading to an increased susceptibility to infection.


Asunto(s)
Regulación hacia Abajo , Productos Finales de Glicación Avanzada/metabolismo , Inmunidad Innata , Inflamasomas/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Activación de Macrófagos/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Organismos Libres de Patógenos Específicos
7.
Int J Neuropsychopharmacol ; 20(11): 896-906, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29016824

RESUMEN

Background: Inflammation may play a significant role in the pathogenesis of depression, although the molecular target for the treatment of inflammation-mediated depressive symptoms remains to be elucidated. Recent studies have implicated the NLRP3 inflammasome in various psychiatric disorders, including depression. However, the underlying mechanism by which NLRP3 inflammasome activation mediates the progression of depressive-like behaviors remains poorly understood. Methods: We examined whether NLRP3 deficiency influenced depressive-like behaviors and cerebral inflammation following systemic administration of lipopolysaccharide in mice. To further assess the contribution of the NLRP3 inflammasome to the progression of depression, we evaluated the effects of NLRP3 signaling on levels of indoleamine 2,3-dioxygenase. Results: Nlrp3-deficient mice exhibited significant attenuation of depressive-like behaviors and cerebral caspase-1 activation in a lipopolysaccharide-induced model of depression. Treatment with the antidepressant amitriptyline failed to block NLRP3-dependent activation of caspase-1, but inhibited lipopolysaccharide-promoted production of interleukin-1ß mRNA via suppressing NF-κB signaling in mouse mixed glial cultures. Interestingly, lipopolysaccharide administration produced NLRP3-dependent increases in indoleamine 2,3-dioxygenase expression and activity of mouse brain. Furthermore, inflammasome-activating stimulations, but not treatment with the inflammasome product interleukin-1ß, triggered indoleamine 2,3-dioxygenase mRNA induction in mixed glial cells. Conclusions: Our data indicate that the NLRP3 inflammasome is significantly implicated in the progression of systemic inflammation-induced depression. NLRP3-dependent caspase-1 activation produced significant increases in indoleamine 2,3-dioxygenase levels, which may play a significant role in lipopolysaccharide-induced depression. Collectively, our findings suggest that indoleamine 2,3-dioxygenase is a potential downstream mediator of the NLRP3 inflammasome in inflammation-mediated depressive-like behaviors.


Asunto(s)
Depresión/inducido químicamente , Depresión/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Lipopolisacáridos/toxicidad , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Amitriptilina/uso terapéutico , Análisis de Varianza , Animales , Encéfalo/citología , Caspasa 1/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Regulación Enzimológica de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , ARN Mensajero
8.
J Biol Chem ; 290(45): 27425-27437, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26416893

RESUMEN

Mitochondrial dysfunction is considered crucial for NLRP3 inflammasome activation partly through its release of mitochondrial toxic products, such as mitochondrial reactive oxygen species (mROS)(2) and mitochondrial DNA (mtDNA). Although previous studies have shown that classical NLRP3-activating stimulations lead to mROS generation and mtDNA release, it remains poorly understood whether and how mitochondrial damage-derived factors may contribute to NLRP3 inflammasome activation. Here, we demonstrate that impairment of the mitochondrial electron transport chain by rotenone primes NLRP3 inflammasome activation only upon costimulation with ATP and not with nigericin or alum. Rotenone-induced priming of NLRP3 in the presence of ATP triggered the formation of specklike NLRP3 or ASC aggregates and the association of NLRP3 with ASC, resulting in NLRP3-dependent caspase-1 activation. Mechanistically, rotenone confers a priming signal for NLRP3 inflammasome activation only in the context of aberrant high-grade, but not low-grade, mROS production and mitochondrial hyperpolarization. By contrast, rotenone/ATP-mediated mtDNA release and mitochondrial depolarization are likely to be merely an indication of mitochondrial damage rather than triggering factors for NLRP3 inflammasome activation. Our results provide a molecular insight into the selective contribution made by mitochondrial dysfunction to the NLRP3 inflammasome pathway.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/antagonistas & inhibidores , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Rotenona/farmacología , Desacopladores/farmacología , Adenosina Trifosfato/metabolismo , Animales , Proteínas Portadoras/genética , Caspasa 1/metabolismo , Células Cultivadas , ADN Mitocondrial/metabolismo , Transporte de Electrón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/metabolismo
9.
Biochem Biophys Res Commun ; 467(4): 973-8, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26471297

RESUMEN

Emerging reports demonstrate that deregulated NLRP3 inflammasome activation is implicated in a variety of inflammatory and metabolic disorders, but the molecular mechanism underlying NLRP3 inflammasome regulation remains uncertain. Here, we present evidence that histone deacetylase 6 (HDAC6) inhibits the activation of NLRP3 inflammasome through its direct association with NLRP3. ShRNA-mediated knockdown of HDAC6 in bone marrow-derived macrophages (BMDMs) showed a significant increase in caspase-1 activation and interleukin-1 beta (IL-1ß) secretion in response to NLRP3-activating stimulations, but not to absent in melanoma 2 (AIM2)-activating stimulation. In addition, knockdown of HDAC6 in BMDMs enhanced the oligomerization of ASC upon LPS/nigericin stimulation. The augmented NLRP3 inflammasome activation seen in HDAC6-knockdown BMDMs is independent of the deacetylase activity of HDAC6. Instead, HDAC6 directly associates with NLRP3 through its ubiquitin-binding domain. Moreover, PR619 treatment (deubiquitinase inhibitor) resulted in the elevation in the interaction of NLRP3 with HDAC6 and the decrease in NLRP3-dependent caspase-1 activation. Taken together, our results indicate that HDAC6 negatively regulates NLRP3 inflammasome activation through its interaction to ubiquitinated NLRP3.


Asunto(s)
Proteínas Portadoras/fisiología , Histona Desacetilasas/metabolismo , Inflamasomas/fisiología , Animales , Sitios de Unión , Células Cultivadas , Histona Desacetilasa 6 , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Transcripción Genética , Ubiquitina/metabolismo
10.
Immunol Cell Biol ; 93(6): 591-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25601272

RESUMEN

Th2 cytokine IL-4 has been previously shown to suppress the production of proinflammatory cytokines in monocytes. However, the underlying molecular mechanism by which IL-4 signaling antagonizes proinflammatory responses is poorly characterized. In particular, whether IL-4 can modulate inflammasome signaling remains unknown. Here, we provide evidence that IL-4 suppresses NLRP3-dependent caspase-1 activation and the subsequent IL-1ß secretion but does not inhibit absent in melanoma 2 (AIM2)- or NLRC4 (NOD-like receptor family, CARD domain-containing 4)-dependent caspase-1 activation in THP-1 and mouse bone marrow-derived macrophages. Upon lipopolysaccharide (LPS) or LPS/ATP stimulation, IL-4 markedly inhibited the assembly of NLRP3 inflammasome, including NLRP3-dependent ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) oligomerization, NLRP3-ASC interaction and NLRP3 speck-like oligomeric structure formation. The negative regulation of NLRP3 inflammasome by IL-4 was not due to the impaired mRNA or protein production of NLRP3 and proinflammatory cytokines. Supporting this observation, IL-4 attenuated NLRP3 inflammasome activation even in reconstituted NLRP3-expressing macrophages in which NLRP3 expression is not transcriptionally regulated by TLR-NF-κB signaling. Furthermore, the IL-4-mediated suppression of NLRP3 inflammasome was independent of STAT6-dependent transcription and mitochondrial reactive oxygen species (ROS). Instead, IL-4 inhibited subcellular redistribution of NLRP3 into mitochondria and microtubule polymerization upon NLRP3-activating stimulation. Our results collectively suggest that IL-4 could suppress NLRP3 inflammasome activation in a transcription-independent manner, thus providing an endogenous regulatory machinery to prevent excessive inflammasome activation.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Interleucina-4/metabolismo , Transducción de Señal , Animales , Caspasa 1/metabolismo , Citocinas/genética , Citocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-4/farmacología , Espacio Intracelular , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Unión Proteica , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
11.
J Immunol ; 191(8): 4358-66, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24048902

RESUMEN

NLRP3 assembles an inflammasome complex that activates caspase-1 upon sensing various danger signals derived from pathogenic infection, tissue damage, and environmental toxins. How NLRP3 senses these various stimuli is still poorly understood, but mitochondria and mitochondrial reactive oxygen species have been proposed to play a critical role in NLRP3 activation. In this article, we provide evidence that the mitochondrial antiviral signaling protein MAVS associates with NLRP3 and facilitates its oligomerization leading to caspase-1 activation. In reconstituted 293T cells, full-length MAVS promoted NLRP3-dependent caspase-1 activation, whereas a C-terminal transmembrane domain-truncated mutant of MAVS (MAVS-ΔTM) did not. MAVS, but not MAVS-ΔTM, interacted with NLRP3 and triggered the oligomerization of NLRP3, suggesting that mitochondrial localization of MAVS and intact MAVS signaling are essential for activating the NLRP3 inflammasome. Supporting this, activation of MAVS signaling by Sendai virus infection promoted NLRP3-dependent caspase-1 activation, whereas knocking down MAVS expression clearly attenuated the activation of NLRP3 inflammasome by Sendai virus in THP-1 and mouse macrophages. Taken together, our results suggest that MAVS facilitates the recruitment of NLRP3 to the mitochondria and may enhance its oligomerization and activation by bringing it in close proximity to mitochondrial reactive oxygen species.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Macrófagos/inmunología , Mitocondrias/metabolismo , Infecciones por Respirovirus/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Caspasa 1/metabolismo , Línea Celular , Activación Enzimática , Células HEK293 , Humanos , Inflamasomas/inmunología , Macrófagos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Interferencia de ARN , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Virus Sendai/inmunología , Transducción de Señal/inmunología
12.
Nature ; 458(7237): 509-13, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19158676

RESUMEN

Host- and pathogen-associated cytoplasmic double-stranded DNA triggers the activation of a NALP3 (also known as cryopyrin and NLRP3)-independent inflammasome, which activates caspase-1 leading to maturation of pro-interleukin-1beta and inflammation. The nature of the cytoplasmic-DNA-sensing inflammasome is currently unknown. Here we show that AIM2 (absent in melanoma 2), an interferon-inducible HIN-200 family member that contains an amino-terminal pyrin domain and a carboxy-terminal oligonucleotide/oligosaccharide-binding domain, senses cytoplasmic DNA by means of its oligonucleotide/oligosaccharide-binding domain and interacts with ASC (apoptosis-associated speck-like protein containing a CARD) through its pyrin domain to activate caspase-1. The interaction of AIM2 with ASC also leads to the formation of the ASC pyroptosome, which induces pyroptotic cell death in cells containing caspase-1. Knockdown of AIM2 by short interfering RNA reduced inflammasome/pyroptosome activation by cytoplasmic DNA in human and mouse macrophages, whereas stable expression of AIM2 in the non-responsive human embryonic kidney 293T cell line conferred responsiveness to cytoplasmic DNA. Our results show that cytoplasmic DNA triggers formation of the AIM2 inflammasome by inducing AIM2 oligomerization. This study identifies AIM2 as an important inflammasome component that senses potentially dangerous cytoplasmic DNA, leading to activation of the ASC pyroptosome and caspase-1.


Asunto(s)
Citoplasma/genética , ADN/metabolismo , Inflamación/metabolismo , Inflamación/patología , Proteínas Nucleares/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/metabolismo , Muerte Celular , Línea Celular , Proteínas del Citoesqueleto/metabolismo , ADN/inmunología , Proteínas de Unión al ADN , Activación Enzimática , Humanos , Ratones , Proteínas Nucleares/química , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Unión Proteica
13.
J Biol Chem ; 288(16): 11378-83, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23479736

RESUMEN

Human pyrin with gain-of-function mutations in its B30.2/SPRY domain causes the autoinflammatory disease familial Mediterranean fever by assembling an ASC-dependent inflammasome that activates caspase-1. Wild-type human pyrin can also form an inflammasome complex with ASC after engagement by autoinflammatory PSTPIP1 mutants. How the pyrin inflammasome is activated in the absence of disease-associated mutations is not yet known. We report here that ribotoxic stress triggers the assembly of the human pyrin inflammasome, leading to ASC oligomerization and caspase-1 activation in THP-1 macrophages and in a 293T cell line stably reconstituted with components of the pyrin inflammasome. Knockdown of pyrin and selective inhibition of p38 MAPK greatly attenuated caspase-1 activation by ribotoxic stress, whereas expression of the conditional mutant ΔMEKK3:ER* allowed the activation of caspase-1 without ribotoxic stress. Disruption of microtubules by colchicine also inhibited pyrin inflammasome activation by ribotoxic stress. Together, our results indicate that ribotoxic stress activates the human pyrin inflammasome through a mechanism that requires p38 MAPK signaling and microtubule stability.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Inflamasomas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Estrés Fisiológico/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular , Colchicina/farmacología , Proteínas del Citoesqueleto/genética , Humanos , Inflamasomas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microtúbulos/genética , Microtúbulos/metabolismo , Mutación , Pirina , Estrés Fisiológico/efectos de los fármacos , Moduladores de Tubulina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
14.
Exp Mol Med ; 56(2): 370-382, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38297162

RESUMEN

Circadian arrhythmia has been linked to increased susceptibility to multiple inflammatory diseases, such as sepsis. However, it remains unclear how disruption of the circadian clock modulates molecular aspects of innate immune responses, including inflammasome signaling. Here, we examined the potential role of the circadian clock in inflammasome-mediated responses through myeloid-specific deletion of BMAL1, a master circadian clock regulator. Intriguingly, Bmal1 deficiency significantly enhanced pyroptosis of macrophages and lethality of mice under noncanonical inflammasome-activating conditions but did not alter canonical inflammasome responses. Transcriptome analysis of enriched peritoneal myeloid cells revealed that Bmal1 deficiency led to a marked reduction in Rev-erbα expression at steady state and a significant increase in serum amyloid A1 (SAA1) expression upon poly(I:C) stimulation. Notably, we found that the circadian regulator Rev-erbα is critical for poly(I:C)- or interferon (IFN)-ß-induced SAA1 production, resulting in the circadian oscillation pattern of SAA1 expression in myeloid cells. Furthermore, exogenously applied SAA1 markedly increased noncanonical inflammasome-mediated pyroptosis of macrophages and lethality of mice. Intriguingly, our results revealed that type 1 IFN receptor signaling is needed for poly(I:C)- or IFN-ß-induced SAA1 production. Downstream of the type 1 IFN receptor, Rev-erbα inhibited the IFN-ß-induced association of C/EBPß with the promoter region of Saa1, leading to the reduced transcription of Saa1 in macrophages. Bmal1-deficient macrophages exhibited enhanced binding of C/EBPß to Saa1. Consistently, the blockade of Rev-erbα by SR8278 significantly increased poly(I:C)-stimulated SAA1 transcription and noncanonical inflammasome-mediated lethality in mice. Collectively, our data demonstrate a potent suppressive effect of the circadian clock BMAL1 on the noncanonical inflammasome response via the Rev-erbα-C/EBPß-SAA1 axis.


Asunto(s)
Relojes Circadianos , Inflamasomas , Animales , Ratones , Factores de Transcripción ARNTL/genética , Relojes Circadianos/genética , Piroptosis , Inmunidad Innata , Proteína beta Potenciadora de Unión a CCAAT/genética , Poli I-C/farmacología
15.
Cell Death Dis ; 15(1): 16, 2024 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184594

RESUMEN

Viruses have evolved to control mitochondrial quality and content to facilitate viral replication. Mitophagy is a selective autophagy, in which the damaged or unnecessary mitochondria are removed, and thus considered an essential mechanism for mitochondrial quality control. Although mitophagy manipulation by several RNA viruses has recently been reported, the effect of mitophagy regulation by varicella zoster virus (VZV) remains to be fully determined. In this study, we showed that dynamin-related protein-1 (DRP1)-mediated mitochondrial fission and subsequent PINK1/Parkin-dependent mitophagy were triggered during VZV infection, facilitating VZV replication. In addition, VZV glycoprotein E (gE) promoted PINK1/Parkin-mediated mitophagy by interacting with LC3 and upregulating mitochondrial reactive oxygen species. Importantly, VZV gE inhibited MAVS oligomerization and STING translocation to disrupt MAVS- and STING-mediated interferon (IFN) responses, and PINK1/Parkin-mediated mitophagy was required for VZV gE-mediated inhibition of IFN production. Similarly, carbonyl cyanide m-chlorophenyl hydrazone (CCCP)-mediated mitophagy induction led to increased VZV replication but attenuated IFN production in a three-dimensional human skin organ culture model. Our results provide new insights into the immune evasion mechanism of VZV gE via PINK1/Parkin-dependent mitophagy.


Asunto(s)
Inmunidad Innata , Mitofagia , Humanos , Carbonil Cianuro m-Clorofenil Hidrazona , Ubiquitina-Proteína Ligasas , Antivirales , Proteínas Quinasas
16.
Arch Pharm Res ; 46(2): 78-89, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36719600

RESUMEN

Inflammation is an essential host defense mechanism in response to microbial infection and tissue injury. In addition to its well-established role in infection, inflammation is actively involved in the repair of damaged tissues and restoration of homeostatic conditions after tissue injury. The intensity of the inflammatory response and types of cells involved in inflammation have a significant impact on the quality of tissue repair. Numerous immune cell subtypes participate in tissue repair and regeneration. In particular, immune cell-derived secretants, including cytokines and growth factors, can actively modulate the proliferation of resident stem cells or progenitor cells to facilitate tissue regeneration. These findings highlight the importance of inflammation during tissue repair and regeneration; however, the precise role of immune cells in tissue regeneration remains unclear. In this review, we summarize the current knowledge on the contribution of specific immune cell types to tissue repair and regeneration. We also discuss how inflammation affects the final outcome of tissue regeneration.


Asunto(s)
Citocinas , Inflamación , Humanos
17.
Immune Netw ; 23(3): e27, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37416934

RESUMEN

Neutrophil extracellular traps (NETs) exert a novel function of trapping pathogens. Released NETs can accumulate in inflamed tissues, be recognized by other immune cells for clearance, and lead to tissue toxicity. Therefore, the deleterious effect of NET is an etiological factor, causing several diseases directly or indirectly. NLR family pyrin domain containing 3 (NLRP3) in neutrophils is pivotal in signaling the innate immune response and is associated with several NET-related diseases. Despite these observations, the role of NLRP3 in NET formation in neuroinflammation remains elusive. Therefore, we aimed to explore NET formation promoted by NLRP3 in an LPS-induced inflamed brain. Wild-type and NLRP3 knockout mice were used to investigate the role of NLRP3 in NET formation. Brain inflammation was systemically induced by administering LPS. In such an environment, the NET formation was evaluated based on the expression of its characteristic indicators. DNA leakage and NET formation were analyzed in both mice through Western blot, flow cytometry, and in vitro live cell imaging as well as two-photon imaging. Our data revealed that NLRP3 promotes DNA leakage and facilitates NET formation accompanied by neutrophil death. Moreover, NLRP3 is not involved in neutrophil infiltration but is predisposed to boost NET formation, which is accompanied by neutrophil death in the LPS-induced inflamed brain. Furthermore, either NLRP3 deficiency or neutrophil depletion diminished pro-inflammatory cytokine, IL-1ß, and alleviated blood-brain barrier damage. Overall, the results suggest that NLRP3 exacerbates NETosis in vitro and in the inflamed brain, aggravating neuroinflammation. These findings provide a clue that NLRP3 would be a potential therapeutic target to alleviate neuroinflammation.

18.
Immune Netw ; 23(3): e23, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37416933

RESUMEN

Inflammation is a series of host defense processes in response to microbial infection and tissue injury. Inflammatory processes frequently cause extracellular acidification in the inflamed region through increased glycolysis and lactate secretion. Therefore, the immune cells infiltrating the inflamed region encounter an acidic microenvironment. Extracellular acidosis can modulate the innate immune response of macrophages; however, its role for inflammasome signaling still remains elusive. In the present study, we demonstrated that macrophages exposed to an acidic microenvironment exhibited enhanced caspase-1 processing and IL-1ß secretion compared with those under physiological pH. Moreover, exposure to an acidic pH increased the ability of macrophages to assemble the NLR family pyrin domain containing 3 (NLRP3) inflammasome in response to an NLRP3 agonist. This acidosis-mediated augmentation of NLRP3 inflammasome activation occurred in bone marrow-derived macrophages but not in bone marrow-derived neutrophils. Notably, exposure to an acidic environment caused a reduction in the intracellular pH of macrophages but not neutrophils. Concordantly, macrophages, but not neutrophils, exhibited NLRP3 agonist-mediated translocation of chloride intracellular channel protein 1 (CLIC1) into their plasma membranes under an acidic microenvironment. Collectively, our results demonstrate that extracellular acidosis during inflammation can increase the sensitivity of NLRP3 inflammasome formation and activation in a CLIC1-dependent manner. Thus, CLIC1 may be a potential therapeutic target for NLRP3 inflammasome-mediated pathological conditions.

19.
J Inflamm Res ; 15: 3053-3063, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35645573

RESUMEN

Background: Peripheral inflammation-triggered mild neuroinflammation impacts the brain and behavior through microglial activation. In this study, we performed an unbiased analysis of the vulnerability of different brain areas to neuroinflammation induced by systemic inflammation. Methods: We injected mice with a single low dose of LPS to induce mild inflammation and then analyzed microglial activation in 34 brain regions by immunohistochemical methods and whole-brain imaging using multi-slide scanning microscopy. We also conducted quantitative RT-PCR to measure the levels of inflammatory cytokines in selected brain regions of interest. Results: We found that microglia in different brain regions are differentially activated by mild, LPS-induced inflammation relative to the increase in microglia numbers or increased CD68 expression. The increased number of microglia induced by mild inflammation was not attributable to infiltration of peripheral immune cells. In addition, microglia residing in brain regions, in which a single low-dose injection of LPS produced microglial changes, preferentially generated pro-inflammatory cytokines. Conclusion: Our results suggest that mild neuroinflammation induces regionally different microglia activation, producing pro-inflammatory cytokines. Our observations provide insight into induction of possible region-specific neuroinflammation-associated brain pathologies through microglial activation.

20.
EBioMedicine ; 82: 104184, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35870427

RESUMEN

BACKGROUND: Cryopyrin-associated periodic syndrome (CAPS) is an inherited autoinflammatory disease caused by a gain-of-function mutation in NLRP3. Although CAPS patients frequently suffer from sensorineural hearing loss, it remains unclear whether CAPS-associated mutation in NLRP3 is associated with the progression of hearing loss. METHODS: We generated a mice with conditional expression of CAPS-associated NLRP3 mutant (D301N) in cochlea-resident CX3CR1 macrophages and examined the susceptibility of CAPS mice to inflammation-mediated hearing loss in a local and systemic inflammation context. FINDINGS: Upon lipopolysaccharide (LPS) injection into middle ear cavity, NLRP3 mutant mice exhibited severe cochlear inflammation, inflammasome activation and hearing loss. However, this middle ear injection model induced a considerable hearing loss in control mice and inevitably caused an inflammation-independent hearing loss possibly due to ear tissue damages by injection procedure. Subsequently, we optimized a systemic LPS injection model, which induced a significant hearing loss in NLRP3 mutant mice but not in control mice. Peripheral inflammation induced by a repetitive low dose of LPS injection caused a blood-labyrinth barrier disruption, macrophage infiltration into cochlea and cochlear inflammasome activation in an NLRP3-dependent manner. Interestingly, both cochlea-infiltrating and -resident macrophages contribute to peripheral inflammation-mediated hearing loss of CAPS mice. Furthermore, NLRP3-specific inhibitor, MCC950, as well as an interleukin-1 receptor antagonist significantly alleviated systemic LPS-induced hearing loss and inflammatory phenotypes in NLRP3 mutant mice. INTERPRETATION: Our findings reveal that CAPS-associated NLRP3 mutation is critical for peripheral inflammation-induced hearing loss in our CAPS mice model, and an NLRP3-specific inhibitor can be used to treat inflammation-mediated sensorineural hearing loss. FUNDING: National Research Foundation of Korea Grant funded by the Korean Government and the Team Science Award of Yonsei University College of Medicine.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina , Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Animales , Síndromes Periódicos Asociados a Criopirina/etiología , Síndromes Periódicos Asociados a Criopirina/genética , Modelos Animales de Enfermedad , Pérdida Auditiva/etiología , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/genética , Humanos , Inflamasomas/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA