Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.406
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 183(4): 982-995.e14, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32991843

RESUMEN

Initially, children were thought to be spared from disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, a month into the epidemic, a novel multisystem inflammatory syndrome in children (MIS-C) emerged. Herein, we report on the immune profiles of nine MIS-C cases. All MIS-C patients had evidence of prior SARS-CoV-2 exposure, mounting an antibody response with intact neutralization capability. Cytokine profiling identified elevated signatures of inflammation (IL-18 and IL-6), lymphocytic and myeloid chemotaxis and activation (CCL3, CCL4, and CDCP1), and mucosal immune dysregulation (IL-17A, CCL20, and CCL28). Immunophenotyping of peripheral blood revealed reductions of non-classical monocytes, and subsets of NK and T lymphocytes, suggesting extravasation to affected tissues. Finally, profiling the autoantigen reactivity of MIS-C plasma revealed both known disease-associated autoantibodies (anti-La) and novel candidates that recognize endothelial, gastrointestinal, and immune-cell antigens. All patients were treated with anti-IL-6R antibody and/or IVIG, which led to rapid disease resolution.


Asunto(s)
Inflamación/patología , Síndrome de Respuesta Inflamatoria Sistémica/patología , Adolescente , Anticuerpos Antivirales/sangre , Autoanticuerpos/sangre , Betacoronavirus/inmunología , Betacoronavirus/aislamiento & purificación , COVID-19 , Quimiocina CCL3/metabolismo , Niño , Preescolar , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Femenino , Humanos , Inmunidad Humoral , Lactante , Recién Nacido , Inflamación/metabolismo , Interleucina-17/metabolismo , Interleucina-18/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Masculino , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/patología , Neumonía Viral/virología , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Adulto Joven
2.
Immunity ; 57(7): 1445-1448, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38986437

RESUMEN

Interleukin-1 (IL-1) family cytokines are key immunological regulators that achieve their signaling prowess after post-translational proteolytic processing. In this issue of Immunity, Dong et al. reveal the structural consequences of this process on proinflammatory IL-18, demonstrating that pro-IL-18 and mature IL-18 are structurally distinct.


Asunto(s)
Interleucina-18 , Transducción de Señal , Interleucina-18/metabolismo , Interleucina-18/inmunología , Humanos , Transducción de Señal/inmunología , Animales , Procesamiento Proteico-Postraduccional
3.
Immunity ; 57(7): 1533-1548.e10, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38733997

RESUMEN

Several interleukin-1 (IL-1) family members, including IL-1ß and IL-18, require processing by inflammasome-associated caspases to unleash their activities. Here, we unveil, by cryoelectron microscopy (cryo-EM), two major conformations of the complex between caspase-1 and pro-IL-18. One conformation is similar to the complex of caspase-4 and pro-IL-18, with interactions at both the active site and an exosite (closed conformation), and the other only contains interactions at the active site (open conformation). Thus, pro-IL-18 recruitment and processing by caspase-1 is less dependent on the exosite than the active site, unlike caspase-4. Structure determination by nuclear magnetic resonance uncovers a compact fold of apo pro-IL-18, which is similar to caspase-1-bound pro-IL-18 but distinct from cleaved IL-18. Binding sites for IL-18 receptor and IL-18 binding protein are only formed upon conformational changes after pro-IL-18 cleavage. These studies show how pro-IL-18 is selected as a caspase-1 substrate, and why cleavage is necessary for its inflammatory activity.


Asunto(s)
Caspasa 1 , Microscopía por Crioelectrón , Interleucina-18 , Transducción de Señal , Interleucina-18/metabolismo , Caspasa 1/metabolismo , Humanos , Inflamasomas/metabolismo , Animales , Conformación Proteica , Unión Proteica , Sitios de Unión , Ratones , Receptores de Interleucina-18/metabolismo , Modelos Moleculares , Péptidos y Proteínas de Señalización Intercelular
4.
Immunity ; 57(9): 2077-2094.e12, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38906145

RESUMEN

Tissues are exposed to diverse inflammatory challenges that shape future inflammatory responses. While cellular metabolism regulates immune function, how metabolism programs and stabilizes immune states within tissues and tunes susceptibility to inflammation is poorly understood. Here, we describe an innate immune metabolic switch that programs long-term intestinal tolerance. Intestinal interleukin-18 (IL-18) stimulation elicited tolerogenic macrophages by preventing their proinflammatory glycolytic polarization via metabolic reprogramming to fatty acid oxidation (FAO). FAO reprogramming was triggered by IL-18 activation of SLC12A3 (NCC), leading to sodium influx, release of mitochondrial DNA, and activation of stimulator of interferon genes (STING). FAO was maintained in macrophages by a bistable switch that encoded memory of IL-18 stimulation and by intercellular positive feedback that sustained the production of macrophage-derived 2'3'-cyclic GMP-AMP (cGAMP) and epithelial-derived IL-18. Thus, a tissue-reinforced metabolic switch encodes durable immune tolerance in the gut and may enable reconstructing compromised immune tolerance in chronic inflammation.


Asunto(s)
Tolerancia Inmunológica , Interleucina-18 , Macrófagos , Nucleótidos Cíclicos , Interleucina-18/metabolismo , Interleucina-18/inmunología , Animales , Ratones , Nucleótidos Cíclicos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Humanos , Ratones Endogámicos C57BL , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones Noqueados , Ácidos Grasos/metabolismo , Intestinos/inmunología , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Glucólisis , Oxidación-Reducción
5.
Nat Immunol ; 21(6): 626-635, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32424362

RESUMEN

The inflammasome NLRP6 plays a crucial role in regulating inflammation and host defense against microorganisms in the intestine. However, the molecular mechanisms by which NLRP6 function is inhibited to prevent excessive inflammation remain unclear. Here, we demonstrate that the deubiquitinase Cyld prevents excessive interleukin 18 (IL-18) production in the colonic mucosa by deubiquitinating NLRP6. We show that deubiquitination inhibited the NLRP6-ASC inflammasome complex and regulated the maturation of IL-18. Cyld deficiency in mice resulted in elevated levels of active IL-18 and severe colonic inflammation following Citrobacter rodentium infection. Further, in patients with ulcerative colitis, the concentration of active IL-18 was inversely correlated with CYLD expression. Thus, we have identified a novel regulatory mechanism that inhibits the NLRP6-IL-18 pathway in intestinal inflammation.


Asunto(s)
Enzima Desubiquitinante CYLD/metabolismo , Enterocolitis/etiología , Enterocolitis/metabolismo , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Citrobacter rodentium , Enzima Desubiquitinante CYLD/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Enterocolitis/patología , Expresión Génica , Humanos , Interleucina-18/antagonistas & inhibidores , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Unión Proteica/inmunología , Ubiquitinación
6.
Immunity ; 55(4): 639-655.e7, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35381213

RESUMEN

Adaptive CD4+ T helper cells and their innate counterparts, innate lymphoid cells, utilize an identical set of transcription factors (TFs) for their differentiation and functions. However, similarities and differences in the induction of these TFs in related lymphocytes are still elusive. Here, we show that T helper-1 (Th1) cells and natural killer (NK) cells displayed distinct epigenomes at the Tbx21 locus, which encodes T-bet, a critical TF for regulating type 1 immune responses. The initial induction of T-bet in NK precursors was dependent on the NK-specific DNase I hypersensitive site Tbx21-CNS-3, and the expression of the interleukin-18 (IL-18) receptor; IL-18 induced T-bet expression through the transcription factor RUNX3, which bound to Tbx21-CNS-3. By contrast, signal transducer and activator of transcription (STAT)-binding motifs within Tbx21-CNS-12 were critical for IL-12-induced T-bet expression during Th1 cell differentiation both in vitro and in vivo. Thus, type 1 innate and adaptive lymphocytes utilize distinct enhancer elements for their development and differentiation.


Asunto(s)
Inmunidad Innata , Interleucina-18 , Células Asesinas Naturales , Células TH1 , Diferenciación Celular , Interleucina-18/metabolismo , Células Asesinas Naturales/inmunología , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Factores de Transcripción/metabolismo
7.
Immunity ; 54(8): 1772-1787.e9, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34289378

RESUMEN

As substantial constituents of the multiple myeloma (MM) microenvironment, pro-inflammatory macrophages have emerged as key promoters of disease progression, bone destruction, and immune impairment. We identify beta-2-microglobulin (ß2m) as a driver in initiating inflammation in myeloma-associated macrophages (MAMs). Lysosomal accumulation of phagocytosed ß2m promotes ß2m amyloid aggregation in MAMs, resulting in lysosomal rupture and ultimately production of active interleukin-1ß (IL-1ß) and IL-18. This process depends on activation of the NLRP3 inflammasome after ß2m accumulation, as macrophages from NLRP3-deficient mice lack efficient ß2m-induced IL-1ß production. Moreover, depletion or silencing of ß2m in MM cells abrogates inflammasome activation in a murine MM model. Finally, we demonstrate that disruption of NLRP3 or IL-18 diminishes tumor growth and osteolytic bone destruction normally promoted by ß2m-induced inflammasome signaling. Our results provide mechanistic evidence for ß2m's role as an NLRP3 inflammasome activator during MM pathogenesis. Moreover, inhibition of NLRP3 represents a potential therapeutic approach in MM.


Asunto(s)
Amiloide/metabolismo , Mieloma Múltiple/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Microglobulina beta-2/metabolismo , Animales , Células Cultivadas , Humanos , Inflamación/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lisosomas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fagocitosis/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Microglobulina beta-2/genética
8.
Nature ; 624(7991): 451-459, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37993712

RESUMEN

Inflammatory caspases are key enzymes in mammalian innate immunity that control the processing and release of interleukin-1 (IL-1)-family cytokines1,2. Despite the biological importance, the structural basis for inflammatory caspase-mediated cytokine processing has remained unclear. To date, catalytic cleavage of IL-1-family members, including pro-IL-1ß and pro-IL-18, has been attributed primarily to caspase-1 activities within canonical inflammasomes3. Here we demonstrate that the lipopolysaccharide receptor caspase-4 from humans and other mammalian species (except rodents) can cleave pro-IL-18 with an efficiency similar to pro-IL-1ß and pro-IL-18 cleavage by the prototypical IL-1-converting enzyme caspase-1. This ability of caspase-4 to cleave pro-IL-18, combined with its previously defined ability to cleave and activate the lytic pore-forming protein gasdermin D (GSDMD)4,5, enables human cells to bypass the need for canonical inflammasomes and caspase-1 for IL-18 release. The structure of the caspase-4-pro-IL-18 complex determined using cryogenic electron microscopy reveals that pro-lL-18 interacts with caspase-4 through two distinct interfaces: a protease exosite and an interface at the caspase-4 active site involving residues in the pro-domain of pro-IL-18, including the tetrapeptide caspase-recognition sequence6. The mechanisms revealed for cytokine substrate capture and cleavage differ from those observed for the caspase substrate GSDMD7,8. These findings provide a structural framework for the discussion of caspase activities in health and disease.


Asunto(s)
Caspasas Iniciadoras , Interleucina-18 , Interleucina-1beta , Animales , Humanos , Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Microscopía por Crioelectrón , Gasderminas/metabolismo , Inflamasomas/metabolismo , Interleucina-18/química , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/metabolismo , Precursores de Proteínas/química , Precursores de Proteínas/metabolismo , Dominio Catalítico
9.
Mol Cell ; 81(22): 4579-4590, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34562371

RESUMEN

Canonically, gasdermin D (GSDMD) cleavage by caspase-1 through inflammasome signaling triggers immune cell pyroptosis (ICP) as a host defense against pathogen infection. However, cancer cell pyroptosis (CCP) was recently discovered to be activated by distinct molecular mechanisms in which GSDMB, GSDMC, and GSDME, rather than GSDMD, are the executioners. Moreover, instead of inflammatory caspases, apoptotic caspases and granzymes are required for gasdermin protein cleavage to induce CCP. Sufficient accumulation of protease-cleaved gasdermin proteins is the prerequisite for CCP. Inflammation induced by ICP or CCP results in diametrically opposite effects on antitumor immunity because of the differential duration and released cellular contents, leading to contrary effects on therapeutic outcomes. Here, we focus on the distinct mechanisms of ICP and CCP and discuss the roles of ICP and CCP in inflammation and antitumor immunity, representing actionable targets.


Asunto(s)
Antineoplásicos/farmacología , Inflamación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Proteínas de Unión a Fosfato/metabolismo , Piroptosis , Animales , Apoptosis , Caspasas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Sistema Inmunológico , Inflamasomas , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/metabolismo , Ratones , Neoplasias/metabolismo , Unión Proteica , Proteolisis , Transducción de Señal
10.
Nat Immunol ; 17(12): 1407-1414, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27798616

RESUMEN

The innate responsiveness of the immune system is important not only for quick responses to pathogens but also for the initiation and shaping of the subsequent adaptive immune response. Activation via the cytokine IL-18, a product of inflammasomes, gives rise to a rapid response that includes the production of self-reactive antibodies. As increased concentrations of this cytokine are found in inflammatory diseases, we investigated the origin of the B cell response and its regulation. We identified an accumulation of B cell-helper neutrophils in the spleen that interacted with innate-type invariant natural killer T cells (iNKT cells) to regulate B cell responses. We found that neutrophil-dependent expression of the death-receptor ligand FasL by iNKT cells was needed to restrict autoantibody production. Neutrophils can thus license iNKT cells to regulate potentially harmful autoreactive B cell responses during inflammasome-driven inflammation.


Asunto(s)
Linfocitos B/inmunología , Proteína Ligando Fas/metabolismo , Inflamación/inmunología , Células T Asesinas Naturales/inmunología , Neutrófilos/inmunología , Inmunidad Adaptativa , Animales , Autoanticuerpos/biosíntesis , Células Cultivadas , Proteína Ligando Fas/genética , Inmunidad Innata , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
EMBO J ; 42(19): e113481, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37575012

RESUMEN

The NLRP3 inflammasome plays a key role in responding to pathogens, and endogenous damage and mitochondria are intensively involved in inflammasome activation. The NLRP3 inflammasome forms multiprotein complexes and its sequential assembly is important for its activation. Here, we show that NLRP3 is ubiquitinated by the mitochondria-associated E3 ligase, MARCH5. Myeloid cell-specific March5 conditional knockout (March5 cKO) mice failed to secrete IL-1ß and IL-18 and exhibited an attenuated mortality rate upon LPS or Pseudomonas aeruginosa challenge. Macrophages derived from March5 cKO mice also did not produce IL-1ß and IL-18 after microbial infection. Mechanistically, MARCH5 interacts with the NACHT domain of NLRP3 and promotes K27-linked polyubiquitination on K324 and K430 residues of NLRP3. Ubiquitination-defective NLRP3 mutants on K324 and K430 residues are not able to bind to NEK7, nor form NLRP3 oligomers leading to abortive ASC speck formation and diminished IL-1ß production. Thus, MARCH5-dependent NLRP3 ubiquitination on the mitochondria is required for NLRP3-NEK7 complex formation and NLRP3 oligomerization. We propose that the E3 ligase MARCH5 is a regulator of NLRP3 inflammasome activation on the mitochondria.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Ratones , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Interleucina-18/metabolismo , Ubiquitinación , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Caspasa 1/metabolismo
12.
Immunity ; 49(3): 413-426.e5, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30170814

RESUMEN

Inflammasome-activated caspase-1 cleaves gasdermin D to unmask its pore-forming activity, the predominant consequence of which is pyroptosis. Here, we report an additional biological role for gasdermin D in limiting cytosolic DNA surveillance. Cytosolic DNA is sensed by Aim2 and cyclic GMP-AMP synthase (cGAS) leading to inflammasome and type I interferon responses, respectively. We found that gasdermin D activated by the Aim2 inflammasome suppressed cGAS-driven type I interferon response to cytosolic DNA and Francisella novicida in macrophages. Similarly, interferon-ß (IFN-ß) response to F. novicida infection was elevated in gasdermin D-deficient mice. Gasdermin D-mediated negative regulation of IFN-ß occurred in a pyroptosis-, interleukin-1 (IL-1)-, and IL-18-independent manner. Mechanistically, gasdermin D depleted intracellular potassium (K+) via membrane pores, and this K+ efflux was necessary and sufficient to inhibit cGAS-dependent IFN-ß response. Thus, our findings have uncovered an additional interferon regulatory module involving gasdermin D and K+ efflux.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Francisella/fisiología , Infecciones por Bacterias Gramnegativas/inmunología , Inflamasomas/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Interferón Tipo I/metabolismo , Interleucina-1/metabolismo , Interleucina-18/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Proteínas de Unión a Fosfato , Potasio/metabolismo , ARN Interferente Pequeño/genética
13.
Immunity ; 49(3): 515-530.e5, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231985

RESUMEN

Fungi represent a significant proportion of the gut microbiota. Aberrant immune responses to fungi are frequently observed in inflammatory bowel diseases (IBD) and colorectal cancer (CRC), and mutations in the fungal-sensing pathways are associated with the pathogenesis of IBD. Fungal recognition receptors trigger downstream signaling via the common adaptor protein CARD9 and the kinase SYK. Here we found that commensal gut fungi promoted inflammasome activation during AOM-DSS-induced colitis. Myeloid cell-specific deletion of Card9 or Syk reduced inflammasome activation and interleukin (IL)-18 maturation and increased susceptibility to colitis and CRC. IL-18 promoted epithelial barrier restitution and interferon-γ production by intestinal CD8+ T cells. Supplementation of IL-18 or transfer of wild-type myeloid cells reduced tumor burden in AOM-DSS-treated Card9-/- and Sykfl/flLysMCre/+ mice, whereas treatment with anti-fungal agents exacerbated colitis and CRC. CARD9 deletion changes the gut microbial landscape, suggesting that SYK-CARD9 signaling maintains a microbial ecology that promotes inflammasome activation and thereby restrains colitis and colon tumorigenesis.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Colitis/inmunología , Neoplasias del Colon/inmunología , Hongos/inmunología , Microbioma Gastrointestinal/inmunología , Inflamasomas/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/fisiología , Células Mieloides/fisiología , Quinasa Syk/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Células Cultivadas , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Humanos , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Dodecil Sulfato de Sodio , Quinasa Syk/genética
14.
Blood ; 144(14): 1496-1507, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-38941593

RESUMEN

ABSTRACT: Kaposi sarcoma herpesvirus (KSHV)-associated diseases include Kaposi sarcoma (KS), primary effusion lymphoma (PEL), KSHV-associated multicentric Castleman disease (MCD), and KS inflammatory cytokine syndrome (KICS). PEL, MCD, and KICS are associated with elevated circulating inflammatory cytokines. However, activation of the inflammasome, which generates interleukin-1ß (IL-1ß) and IL-18 via active caspase-1/4/5, has not been evaluated in patients with KSHV-associated diseases (KADs). Herein we report that patients with HIV and ≥1 KAD present with higher plasma levels of IL-18 and increased caspase-1/4/5 activity in circulating monocytes compared with HIV-negative healthy volunteers (HVs) or people with HIV (PWH) without KAD. Within KAD subtypes, KICS and MCD shared enhanced caspase-1/4/5 activity and IL-18 production compared with HVs and PWH, whereas patients with PEL showed remarkably high levels of inflammasome complex formation (known as apoptosis-associated speck-like protein containing a caspase recruitment domain). Moreover, caspase-1/4/5 activity and IL-18 plasma levels correlated with KSHV viral load, indicating KSHV-driven inflammasome activation in KAD. Accordingly, factors released by cells latently infected with KSHV triggered inflammasome activation and cytokine production in bystander monocytes in vitro. Finally, both supervised and unsupervised analyses with inflammasome measurements and other inflammatory biomarkers demonstrate a unique inflammatory profile in patients with PEL, MCD, and KICS as compared with KS. Our data indicate that detrimental inflammation in patients with KAD is at least partially driven by KSHV-induced inflammasome activation in monocytes, thus offering novel approaches to diagnose and treat these complex disorders. These trials were registered at www.ClinicalTrials.gov as #NCT01419561, NCT00092222, NCT00006518, and NCT02147405.


Asunto(s)
Enfermedad de Castleman , Herpesvirus Humano 8 , Inflamasomas , Sarcoma de Kaposi , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Caspasas/metabolismo , Enfermedad de Castleman/virología , Enfermedad de Castleman/inmunología , Enfermedad de Castleman/sangre , Herpesvirus Humano 8/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , Infecciones por VIH/sangre , Inflamasomas/metabolismo , Inflamasomas/inmunología , Interleucina-18/sangre , Interleucina-18/metabolismo , Linfoma de Efusión Primaria/virología , Linfoma de Efusión Primaria/inmunología , Monocitos/metabolismo , Monocitos/inmunología , Sarcoma de Kaposi/virología , Sarcoma de Kaposi/inmunología , Sarcoma de Kaposi/sangre
15.
Immunity ; 46(4): 649-659, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28410991

RESUMEN

Intestinal epithelial cells (IECs) form a critical barrier against pathogen invasion. By generation of mice in which inflammasome expression is restricted to IECs, we describe a coordinated epithelium-intrinsic inflammasome response in vivo. This response was sufficient to protect against Salmonella tissue invasion and involved a previously reported IEC expulsion that was coordinated with lipid mediator and cytokine production and lytic IEC death. Excessive inflammasome activation in IECs was sufficient to result in diarrhea and pathology. Experiments with IEC organoids demonstrated that IEC expulsion did not require other cell types. IEC expulsion was accompanied by a major actin rearrangement in neighboring cells that maintained epithelium integrity but did not absolutely require Caspase-1 or Gasdermin D. Analysis of Casp1-/-Casp8-/- mice revealed a functional Caspase-8 inflammasome in vivo. Thus, a coordinated IEC-intrinsic, Caspase-1 and -8 inflammasome response plays a key role in intestinal immune defense and pathology.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Caspasa 1/metabolismo , Caspasa 8/metabolismo , Eicosanoides/metabolismo , Células Epiteliales/metabolismo , Interleucina-18/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Caspasa 1/genética , Caspasa 8/genética , Activación Enzimática , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/microbiología , Inflamasomas/genética , Inflamasomas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Péptidos y Proteínas de Señalización Intracelular , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Proteínas de Unión a Fosfato , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/fisiología
16.
J Immunol ; 212(4): 596-606, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38149914

RESUMEN

Inflammasome-mediated caspase-1 activation facilitates innate immune control of Plasmodium in the liver, thereby limiting the incidence and severity of clinical malaria. However, caspase-1 processing occurs incompletely in both mouse and human hepatocytes and precludes the generation of mature IL-1ß or IL-18, unlike in other cells. Why this is so or how it impacts Plasmodium control in the liver has remained unknown. We show that an inherently reduced expression of the inflammasome adaptor molecule apoptosis-associated specklike protein containing CARD (ASC) is responsible for the incomplete proteolytic processing of caspase-1 in murine hepatocytes. Transgenically enhancing ASC expression in hepatocytes enabled complete caspase-1 processing, enhanced pyroptotic cell death, maturation of the proinflammatory cytokines IL-1ß and IL-18 that was otherwise absent, and better overall control of Plasmodium infection in the liver of mice. This, however, impeded the protection offered by live attenuated antimalarial vaccination. Tempering ASC expression in mouse macrophages, on the other hand, resulted in incomplete processing of caspase-1. Our work shows how caspase-1 activation and function in host cells are fundamentally defined by ASC expression and offers a potential new pathway to create better disease and vaccination outcomes by modifying the latter.


Asunto(s)
Inflamasomas , Malaria , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Hepatocitos/metabolismo , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
17.
Eur J Immunol ; 54(10): e2451053, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39072707

RESUMEN

Cigarette smoke (CS) is a major risk factor for chronic lung diseases and promotes activation of pattern recognition receptors in the bronchial epithelium. NOD-like receptor family, pyrin domain-containing 3 (NLRP3) is a pattern recognition receptor whose activation leads to caspase-1 cleavage, maturation/release of IL-1ß and IL-18, and eventually pyroptosis. Whether the NLRP3 inflammasome participates in CS-induced inflammation in bronchial epithelial cells is still unclear. Herein, we evaluated the involvement of NLRP3 in CS-induced inflammatory responses in human primary bronchial epithelial cells. To this purpose, human primary bronchial epithelial cells were stimulated with CS extracts (CSE) and lytic cell death, caspase activation (-1, -8, -3/7), cytokine release (IL-1ß, IL-18, and IL-8), NLRP3, pro-IL-1ß/pro-IL-18 mRNA, and protein expression were measured. The impact of inhibitors of NLRP3 (MCC950), caspases, and the effect of the antioxidant N-acetyl cysteine were evaluated. We found that CSE increased pro-IL-1ß expression and induced activation of caspase-1 and release of IL-1ß and IL-18. These events were independent of NLRP3 and we found that NLRP3 was not expressed. N-acetyl cysteine reverted CSE-induced caspase-1 activation. Overall, our findings support that the bronchial epithelium may play a central role in the release of IL-1 family cytokines independently of NLRP3 in the lungs of smokers.


Asunto(s)
Bronquios , Células Epiteliales , Inflamasomas , Interleucina-18 , Interleucina-1beta , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Células Epiteliales/metabolismo , Bronquios/citología , Bronquios/patología , Bronquios/metabolismo , Inflamasomas/metabolismo , Células Cultivadas , Humo/efectos adversos , Caspasa 1/metabolismo , Fumar Cigarrillos/efectos adversos
18.
PLoS Pathog ; 19(11): e1011747, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37910490

RESUMEN

Buruli ulcer is an emerging chronic infectious skin disease caused by Mycobacterium ulcerans. Mycolactone, an exotoxin produced by the bacterium, is the only identified virulence factor so far, but the functions of this toxin and the mechanisms of disease progression remain unclear. By interfering Sec61 translocon, mycolactone inhibits the Sec61-dependent co-translational translocation of newly synthesized proteins, such as induced cytokines and immune cell receptors, into the endoplasmic reticulum. However, in regard to IL-1ß, which is secreted by a Sec61-independent mechanism, mycolactone has been shown to induce IL-1ß secretion via activation of inflammasomes. In this study, we clarified that cytokine induction, including that of IL-1ß, in infected macrophages was suppressed by mycolactone produced by M. ulcerans subsp. shinshuense, despite the activation of caspase-1 through the inflammasome activation triggered in a manner independent of mycolactone. Intriguingly, mycolactone suppressed the expression of proIL-1ß as well as TNF-α at the transcriptional level, suggesting that mycolactone of M. ulcerans subsp. shinshuense may exert additional inhibitory effect on proIL-1ß expression. Remarkably, constitutively produced IL-18 was cleaved and mature IL-18 was actually released from macrophages infected with the causative mycobacterium. IL-18-deficient mice infected subcutaneously with M. ulcerans exhibited exacerbated skin inflammation during the course of disease progression. On the other hand, IL-1ß controls bacterial multiplication in skin tissues. These results provide information regarding the mechanisms and functions of the induced cytokines in the pathology of Buruli ulcer.


Asunto(s)
Úlcera de Buruli , Mycobacterium ulcerans , Animales , Ratones , Úlcera de Buruli/microbiología , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Mycobacterium ulcerans/metabolismo , Macrólidos/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Inflamación
19.
PLoS Pathog ; 19(4): e1011321, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37068092

RESUMEN

Group A Streptococcus (GAS, Streptococcus pyogenes) is a professional human pathogen that commonly infects the skin. Keratinocytes are one of the first cells to contact GAS, and by inducing inflammation, they can initiate the earliest immune responses to pathogen invasion. Here, we characterized the proinflammatory cytokine repertoire produced by primary human keratinocytes and surrogate cell lines commonly used in vitro. Infection induces several cytokines and chemokines, but keratinocytes constitutively secrete IL-18 in a form that is inert (pro-IL-18) and lacks proinflammatory activity. Canonically, IL-18 activation and secretion are coupled through a single proteolytic event that is regulated intracellularly by the inflammasome protease caspase-1 in myeloid cells. The pool of extracellular pro-IL-18 generated by keratinocytes is poised to sense extracellular proteases. It is directly processed into a mature active form by SpeB, a secreted GAS protease that is a critical virulent factor during skin infection. This mechanism contributes to the proinflammatory response against GAS, resulting in T cell activation and the secretion of IFN-γ. Under these conditions, isolates of several other major bacterial pathogens and microbiota of the skin were found to not have significant IL-18-maturing ability. These results suggest keratinocyte-secreted IL-18 is a sentinel that sounds an early alarm that is highly sensitive to GAS, yet tolerant to non-invasive members of the microbiota.


Asunto(s)
Infecciones Bacterianas , Interleucina-18 , Humanos , Infecciones Bacterianas/metabolismo , Citocinas/metabolismo , Inflamación , Interleucina-18/metabolismo , Queratinocitos/metabolismo , Péptido Hidrolasas/metabolismo
20.
Nat Immunol ; 14(5): 480-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23525089

RESUMEN

NOD2 receptor and the cytosolic protein kinase RIPK2 regulate NF-κB and MAP kinase signaling during bacterial infections, but the role of this immune axis during viral infections has not been addressed. We demonstrate that Nod2(-/-) and Ripk2(-/-) mice are hypersusceptible to infection with influenza A virus. Ripk2(-/-) cells exhibited defective autophagy of mitochondria (mitophagy), leading to enhanced mitochondrial production of superoxide and accumulation of damaged mitochondria, which resulted in greater activation of the NLRP3 inflammasome and production of IL-18. RIPK2 regulated mitophagy in a kinase-dependent manner by phosphorylating the mitophagy inducer ULK1. Accordingly, Ulk1(-/-) cells exhibited enhanced mitochondrial production of superoxide and activation of caspase-1. These results demonstrate a role for NOD2-RIPK2 signaling in protection against virally triggered immunopathology by negatively regulating activation of the NLRP3 inflammasome and production of IL-18 via ULK1-dependent mitophagy.


Asunto(s)
Alphainfluenzavirus/inmunología , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Mitocondrias/fisiología , Mitofagia , Infecciones por Orthomyxoviridae/inmunología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia , Caspasa 1/metabolismo , Células Cultivadas , Inmunidad Activa/genética , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Transducción de Señal/genética , Superóxidos/metabolismo , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA