Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(22): eadl0320, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38820160

RESUMEN

Translation of mRNAs is a fundamental process that occurs in all cell types of multicellular organisms. Conventionally, it has been considered a default step in gene expression, lacking specific regulation. However, recent studies have documented that certain mRNAs exhibit cell type-specific translation. Despite this, it remains unclear whether global translation is controlled in a cell type-specific manner. By using human cell lines and mouse models, we found that deletion of the ribosome-associated protein ribonuclease inhibitor 1 (RNH1) decreases global translation selectively in hematopoietic-origin cells but not in the non-hematopoietic-origin cells. RNH1-mediated cell type-specific translation is mechanistically linked to angiogenin-induced ribosomal biogenesis. Collectively, this study unravels the existence of cell type-specific global translation regulators and highlights the complex translation regulation in vertebrates.


Asunto(s)
Biosíntesis de Proteínas , Ribonucleasa Pancreática , Ribosomas , Ribonucleasa Pancreática/metabolismo , Ribonucleasa Pancreática/genética , Humanos , Animales , Ratones , Ribosomas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación de la Expresión Génica , Línea Celular , Especificidad de Órganos , Proteínas Portadoras
2.
Life Sci Alliance ; 5(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35256513

RESUMEN

Inflammasomes are cytosolic innate immune sensors of pathogen infection and cellular damage that induce caspase-1-mediated inflammation upon activation. Although inflammation is protective, uncontrolled excessive inflammation can cause inflammatory diseases and can be detrimental, such as in coronavirus disease (COVID-19). However, the underlying mechanisms that control inflammasome activation are incompletely understood. Here we report that the leucine-rich repeat (LRR) protein ribonuclease inhibitor (RNH1), which shares homology with LRRs of NLRP (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing) proteins, attenuates inflammasome activation. Deletion of RNH1 in macrophages increases interleukin (IL)-1ß production and caspase-1 activation in response to inflammasome stimulation. Mechanistically, RNH1 decreases pro-IL-1ß expression and induces proteasome-mediated caspase-1 degradation. Corroborating this, mouse models of monosodium urate (MSU)-induced peritonitis and lipopolysaccharide (LPS)-induced endotoxemia, which are dependent on caspase-1, respectively, show increased neutrophil infiltration and lethality in Rnh1 -/- mice compared with wild-type mice. Furthermore, RNH1 protein levels were negatively related with disease severity and inflammation in hospitalized COVID-19 patients. We propose that RNH1 is a new inflammasome regulator with relevance to COVID-19 severity.


Asunto(s)
COVID-19/patología , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Proteínas Repetidas Ricas en Leucina/metabolismo , Animales , COVID-19/inmunología , Caspasa 1/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Gravedad del Paciente , Complejo de la Endopetidasa Proteasomal/metabolismo
3.
Nat Commun ; 9(1): 1199, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29572442

RESUMEN

The B cell survival factor (TNFSF13B/BAFF) is often elevated in autoimmune diseases and is targeted in the clinic for the treatment of systemic lupus erythematosus. BAFF contains a loop region designated the flap, which is dispensable for receptor binding. Here we show that the flap of BAFF has two functions. In addition to facilitating the formation of a highly active BAFF 60-mer as shown previously, it also converts binding of BAFF to TNFRSF13C (BAFFR) into a signaling event via oligomerization of individual BAFF-BAFFR complexes. Binding and activation of BAFFR can therefore be targeted independently to inhibit or activate the function of BAFF. Moreover, structural analyses suggest that the flap of BAFF 60-mer temporarily prevents binding of an anti-BAFF antibody (belimumab) but not of a decoy receptor (atacicept). The observed differences in profiles of BAFF inhibition may confer distinct biological and clinical efficacies to these therapeutically relevant inhibitors.


Asunto(s)
Factor Activador de Células B/química , Factor Activador de Células B/fisiología , Receptor del Factor Activador de Células B/química , Linfocitos B/citología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Factor Activador de Células B/genética , Diferenciación Celular , Supervivencia Celular , Reactivos de Enlaces Cruzados/química , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Fragmentos de Inmunoglobulinas/química , Linfopenia/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes de Fusión/farmacología
4.
J Clin Invest ; 128(4): 1597-1614, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29408805

RESUMEN

Ribosomal proteins (RP) regulate specific gene expression by selectively translating subsets of mRNAs. Indeed, in Diamond-Blackfan anemia and 5q- syndrome, mutations in RP genes lead to a specific defect in erythroid gene translation and cause anemia. Little is known about the molecular mechanisms of selective mRNA translation and involvement of ribosomal-associated factors in this process. Ribonuclease inhibitor 1 (RNH1) is a ubiquitously expressed protein that binds to and inhibits pancreatic-type ribonucleases. Here, we report that RNH1 binds to ribosomes and regulates erythropoiesis by controlling translation of the erythroid transcription factor GATA1. Rnh1-deficient mice die between embryonic days E8.5 and E10 due to impaired production of mature erythroid cells from progenitor cells. In Rnh1-deficient embryos, mRNA levels of Gata1 are normal, but GATA1 protein levels are decreased. At the molecular level, we found that RNH1 binds to the 40S subunit of ribosomes and facilitates polysome formation on Gata1 mRNA to confer transcript-specific translation. Further, RNH1 knockdown in human CD34+ progenitor cells decreased erythroid differentiation without affecting myelopoiesis. Our results reveal an unsuspected role for RNH1 in the control of GATA1 mRNA translation and erythropoiesis.


Asunto(s)
Embrión de Mamíferos/metabolismo , Eritropoyesis , Factor de Transcripción GATA1/biosíntesis , Células Madre Hematopoyéticas/metabolismo , Biosíntesis de Proteínas , Proteínas/metabolismo , Animales , Embrión de Mamíferos/citología , Factor de Transcripción GATA1/genética , Células Madre Hematopoyéticas/citología , Humanos , Células K562 , Ratones , Ratones Noqueados , Proteínas/genética , Subunidades Ribosómicas Grandes/genética , Subunidades Ribosómicas Grandes/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(32): E4671-80, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27462105

RESUMEN

Inflammasomes are critical sensors that convey cellular stress and pathogen presence to the immune system by activating inflammatory caspases and cytokines such as IL-1ß. The nature of endogenous stress signals that activate inflammasomes remains unclear. Here we show that an inhibitor of the HIV aspartyl protease, Nelfinavir, triggers inflammasome formation and elicits an IL-1R-dependent inflammation in mice. We found that Nelfinavir impaired the maturation of lamin A, a structural component of the nuclear envelope, thereby promoting the release of DNA in the cytosol. Moreover, deficiency of the cytosolic DNA-sensor AIM2 impaired Nelfinavir-mediated inflammasome activation. These findings identify a pharmacologic activator of inflammasome and demonstrate the role of AIM2 in detecting endogenous DNA release upon perturbation of nuclear envelope integrity.


Asunto(s)
Inflamasomas/efectos de los fármacos , Nelfinavir/farmacología , Membrana Nuclear/efectos de los fármacos , Animales , Proteínas Adaptadoras de Señalización CARD/fisiología , Caspasa 1/metabolismo , ADN/metabolismo , Inflamasomas/fisiología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Membrana Nuclear/fisiología , Receptores de Interleucina-1/fisiología
6.
J Biol Chem ; 291(38): 19826-34, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27451394

RESUMEN

B cell activating factor of the TNF family (BAFF), also known as B lymphocyte stimulator, is a ligand required for the generation and maintenance of B lymphocytes. In this study, the ability of different monoclonal antibodies to recognize, inhibit, or activate mouse BAFF was investigated. One of them, a mouse IgG1 named Sandy-2, prevented the binding of BAFF to all of its receptors, BAFF receptor, transmembrane activator and calcium modulating ligand interactor, and B cell maturation antigen, at a stoichiometric ratio; blocked the activity of mouse BAFF on a variety of cell-based reporter assays; and antagonized the prosurvival action of BAFF on primary mouse B cells in vitro A single administration of Sandy-2 in mice induced B cell depletion within 2 weeks, down to levels close to those observed in BAFF-deficient mice. This depletion could then be maintained with a chronic treatment. Sandy-2 and a previously described rat IgG1 antibody, 5A8, also formed a pair suitable for the sensitive detection of endogenous circulating BAFF by ELISA or using a homogenous assay. Interestingly, 5A8 and Sandy-5 displayed activities opposite to that of Sandy-2 by stimulating recombinant BAFF in vitro and endogenous BAFF in vivo These tools will prove useful for the detection and functional manipulation of endogenous mouse BAFF and provide an alternative to the widely used BAFF receptor-Fc decoy receptor for the specific depletion of BAFF in mice.


Asunto(s)
Anticuerpos/farmacología , Factor Activador de Células B/antagonistas & inhibidores , Linfocitos B/inmunología , Inmunoglobulina G/farmacología , Animales , Anticuerpos/inmunología , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Linfocitos B/patología , Supervivencia Celular/efectos de los fármacos , Hiperplasia , Inmunoglobulina G/inmunología , Depleción Linfocítica/métodos , Ratones , Ratones Noqueados
7.
J Immunol ; 196(7): 2939-46, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26944927

RESUMEN

NLRC5, a member of the NOD-like receptor (NLR) protein family, has recently been characterized as the master transcriptional regulator of MHCI molecules in lymphocytes, in which it is highly expressed. However, its role in activated dendritic cells (DCs), which are instrumental to initiate T cell responses, remained elusive. We show in this study that, following stimulation of DCs with inflammatory stimuli, not only did NLRC5 level increase, but also its importance in directing MHCI transcription. Despite markedly reduced mRNA and intracellular H2-K levels, we unexpectedly observed nearly normal H2-K surface display in Nlrc5(-/-) DCs. Importantly, this discrepancy between a strong intracellular and a mild surface defect in H2-K levels was observed also in DCs with H2-K transcription defects independent of Nlrc5. Hence, alongside with demonstrating the importance of NLRC5 in MHCI transcription in activated DCs, we uncover a general mechanism counteracting low MHCI surface expression. In agreement with the decreased amount of neosynthesized MHCI, Nlrc5(-/-) DCs exhibited a defective capacity to display endogenous Ags. However, neither T cell priming by endogenous Ags nor cross-priming ability was substantially affected in activated Nlrc5(-/-) DCs. Altogether, these data show that Nlrc5 deficiency, despite significantly affecting MHCI transcription and Ag display, is not sufficient to hinder T cell activation, underlining the robustness of the T cell priming process by activated DCs.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Animales , Presentación de Antígeno/inmunología , Línea Celular , Membrana Celular/metabolismo , Reactividad Cruzada/inmunología , Regulación de la Expresión Génica , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Linfocitos T/metabolismo , Transcripción Genética
8.
PLoS One ; 10(6): e0130624, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26091541

RESUMEN

Neuroinflammation is the local reaction of the brain to infection, trauma, toxic molecules or protein aggregates. The brain resident macrophages, microglia, are able to trigger an appropriate response involving secretion of cytokines and chemokines, resulting in the activation of astrocytes and recruitment of peripheral immune cells. IL-1ß plays an important role in this response; yet its production and mode of action in the brain are not fully understood and its precise implication in neurodegenerative diseases needs further characterization. Our results indicate that the capacity to form a functional NLRP3 inflammasome and secretion of IL-1ß is limited to the microglial compartment in the mouse brain. We were not able to observe IL-1ß secretion from astrocytes, nor do they express all NLRP3 inflammasome components. Microglia were able to produce IL-1ß in response to different classical inflammasome activators, such as ATP, Nigericin or Alum. Similarly, microglia secreted IL-18 and IL-1α, two other inflammasome-linked pro-inflammatory factors. Cell stimulation with α-synuclein, a neurodegenerative disease-related peptide, did not result in the release of active IL-1ß by microglia, despite a weak pro-inflammatory effect. Amyloid-ß peptides were able to activate the NLRP3 inflammasome in microglia and IL-1ß secretion occurred in a P2X7 receptor-independent manner. Thus microglia-dependent inflammasome activation can play an important role in the brain and especially in neuroinflammatory conditions.


Asunto(s)
Encéfalo/citología , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Microglía/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Astrocitos/metabolismo , Proteínas Portadoras/genética , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/metabolismo , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Interleucina-18/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/análisis , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Microglía/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR , Fragmentos de Péptidos/toxicidad , Receptores Purinérgicos P2X7/metabolismo , alfa-Sinucleína/farmacología
9.
J Biol Chem ; 290(26): 16330-42, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25953898

RESUMEN

The closely related TNF family ligands B cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL) serve in the generation and maintenance of mature B-lymphocytes. Both BAFF and APRIL assemble as homotrimers that bind and activate several receptors that they partially share. However, heteromers of BAFF and APRIL that occur in patients with autoimmune diseases are incompletely characterized. The N and C termini of adjacent BAFF or APRIL monomers are spatially close and can be linked to create single-chain homo- or hetero-ligands of defined stoichiometry. Similar to APRIL, heteromers consisting of one BAFF and two APRILs (BAA) bind to the receptors B cell maturation antigen (BCMA), transmembrane activator and CAML interactor (TACI) but not to the BAFF receptor (BAFFR). Heteromers consisting of one APRIL and two BAFF (ABB) bind to TACI and BCMA and weakly to BAFFR in accordance with the analysis of the receptor interaction sites in the crystallographic structure of ABB. Receptor binding correlated with activity in reporter cell line assays specific for BAFFR, TACI, or BCMA. Single-chain BAFF (BBB) and to a lesser extent single-chain ABB, but not APRIL or single-chain BAA, rescued BAFFR-dependent B cell maturation in BAFF-deficient mice. In conclusion, BAFF-APRIL heteromers of different stoichiometries have distinct receptor-binding properties and activities. Based on the observation that heteromers are less active than BAFF, we speculate that their physiological role might be to down-regulate BAFF activity.


Asunto(s)
Factor Activador de Células B/metabolismo , Antígeno de Maduración de Linfocitos B/metabolismo , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Factor Activador de Células B/química , Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/metabolismo , Antígeno de Maduración de Linfocitos B/genética , Dimerización , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Transducción de Señal , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/química , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
10.
J Exp Med ; 212(3): 369-83, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25732303

RESUMEN

NLR family apoptosis inhibitory proteins (NAIPs) belong to both the Nod-like receptor (NLR) and the inhibitor of apoptosis (IAP) families. NAIPs are known to form an inflammasome with NLRC4, but other in vivo functions remain unexplored. Using mice deficient for all NAIP paralogs (Naip1-6(Δ/Δ)), we show that NAIPs are key regulators of colorectal tumorigenesis. Naip1-6(Δ/Δ) mice developed increased colorectal tumors, in an epithelial-intrinsic manner, in a model of colitis-associated cancer. Increased tumorigenesis, however, was not driven by an exacerbated inflammatory response. Instead, Naip1-6(Δ/Δ) mice were protected from severe colitis and displayed increased antiapoptotic and proliferation-related gene expression. Naip1-6(Δ/Δ) mice also displayed increased tumorigenesis in an inflammation-independent model of colorectal cancer. Moreover, Naip1-6(Δ/Δ) mice, but not Nlrc4-null mice, displayed hyper-activation of STAT3 and failed to activate p53 18 h after carcinogen exposure. This suggests that NAIPs protect against tumor initiation in the colon by promoting the removal of carcinogen-elicited epithelium, likely in a NLRC4 inflammasome-independent manner. Collectively, we demonstrate a novel epithelial-intrinsic function of NAIPs in protecting the colonic epithelium against tumorigenesis.


Asunto(s)
Colitis/patología , Neoplasias del Colon/patología , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Animales , Colitis/genética , Colitis/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Inflamasomas/genética , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Inhibidora de la Apoptosis Neuronal/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
11.
J Immunol ; 194(2): 499-503, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25505286

RESUMEN

Myocardial infarction (MI) induces a sterile inflammatory response that contributes to adverse cardiac remodeling. The initiating mechanisms of this response remain incompletely defined. We found that necrotic cardiomyocytes released a heat-labile proinflammatory signal activating MAPKs and NF-κB in cardiac fibroblasts, with secondary production of cytokines. This response was abolished in Myd88(-/-) fibroblasts but was unaffected in nlrp3-deficient fibroblasts. Despite MyD88 dependency, the response was TLR independent, as explored in TLR reporter cells, pointing to a contribution of the IL-1 pathway. Indeed, necrotic cardiomyocytes released IL-1α, but not IL-1ß, and the immune activation of cardiac fibroblasts was abrogated by an IL-1R antagonist and an IL-1α-blocking Ab. Moreover, immune responses triggered by necrotic Il1a(-/-) cardiomyocytes were markedly reduced. In vivo, mice exposed to MI released IL-1α in the plasma, and postischemic inflammation was attenuated in Il1a(-/-) mice. Thus, our findings identify IL-1α as a crucial early danger signal triggering post-MI inflammation.


Asunto(s)
Interleucina-1alfa/inmunología , Infarto del Miocardio/inmunología , Miocarditis/inmunología , Miocitos Cardíacos/inmunología , Transducción de Señal/inmunología , Animales , Inflamación/etiología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1alfa/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocarditis/etiología , Miocarditis/genética , Miocarditis/patología , Miocitos Cardíacos/patología , Transducción de Señal/genética , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
12.
Proc Natl Acad Sci U S A ; 111(48): 17254-9, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25404286

RESUMEN

Inflammasomes are caspase-1-activating multiprotein complexes. The mouse nucleotide-binding domain and leucine rich repeat pyrin containing 1b (NLRP1b) inflammasome was identified as the sensor of Bacillus anthracis lethal toxin (LT) in mouse macrophages from sensitive strains such as BALB/c. Upon exposure to LT, the NLRP1b inflammasome activates caspase-1 to produce mature IL-1ß and induce pyroptosis. Both processes are believed to depend on autoproteolysed caspase-1. In contrast to human NLRP1, mouse NLRP1b lacks an N-terminal pyrin domain (PYD), indicating that the assembly of the NLRP1b inflammasome does not require the adaptor apoptosis-associated speck-like protein containing a CARD (ASC). LT-induced NLRP1b inflammasome activation was shown to be impaired upon inhibition of potassium efflux, which is known to play a major role in NLRP3 inflammasome formation and ASC dimerization. We investigated whether NLRP3 and/or ASC were required for caspase-1 activation upon LT stimulation in the BALB/c background. The NLRP1b inflammasome activation was assessed in both macrophages and dendritic cells lacking either ASC or NLRP3. Upon LT treatment, the absence of NLRP3 did not alter the NLRP1b inflammasome activity. Surprisingly, the absence of ASC resulted in IL-1ß cleavage and pyroptosis, despite the absence of caspase-1 autoprocessing activity. By reconstituting caspase-1/caspase-11(-/-) cells with a noncleavable or catalytically inactive mutant version of caspase-1, we directly demonstrated that noncleavable caspase-1 is fully active in response to the NLRP1b activator LT, whereas it is nonfunctional in response to the NLRP3 activator nigericin. Taken together, these results establish variable requirements for caspase-1 cleavage depending on the pathogen and the responding NLR.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Western Blotting , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Caspasa 1/genética , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Inflamasomas/genética , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Modelos Biológicos , Proteína con Dominio Pirina 3 de la Familia NLR , Nigericina/farmacología , Proteolisis
13.
Cell Host Microbe ; 16(2): 237-248, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25121751

RESUMEN

The gut mucosal epithelium separates the host from the microbiota, but enteropathogens such as Salmonella Typhimurium (S.Tm) can invade and breach this barrier. Defenses against such acute insults remain incompletely understood. Using a murine model of Salmonella enterocolitis, we analyzed mechanisms limiting pathogen loads in the epithelium during early infection. Although the epithelium-invading S.Tm replicate initially, this intraepithelial replicative niche is restricted by expulsion of infected enterocytes into the lumen. This mechanism is compromised if inflammasome components (NAIP1-6, NLRC4, caspase-1/-11) are deleted, or ablated specifically in the epithelium, resulting in ∼100-fold higher intraepithelial loads and accelerated lymph node colonization. Interestingly, the cytokines downstream of inflammasome activation, interleukin (IL)-1α/ß and IL-18, appear dispensable for epithelial restriction of early infection. These data establish the role of an epithelium-intrinsic inflammasome, which drives expulsion of infected cells to restrict the pathogen's intraepithelial proliferation. This may represent a general defense mechanism against mucosal infections.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al Calcio/fisiología , Enterocitos/microbiología , Proteína Inhibidora de la Apoptosis Neuronal/fisiología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Animales , Ciego/microbiología , Ciego/patología , Enterocitos/inmunología , Interacciones Huésped-Patógeno , Inflamasomas , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones Endogámicos C57BL , Ratones Noqueados
14.
J Biol Chem ; 289(7): 4273-85, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24391090

RESUMEN

Development of ectodermal appendages, such as hair, teeth, sweat glands, sebaceous glands, and mammary glands, requires the action of the TNF family ligand ectodysplasin A (EDA). Mutations of the X-linked EDA gene cause reduction or absence of many ectodermal appendages and have been identified as a cause of ectodermal dysplasia in humans, mice, dogs, and cattle. We have generated blocking antibodies, raised in Eda-deficient mice, against the conserved, receptor-binding domain of EDA. These antibodies recognize epitopes overlapping the receptor-binding site and prevent EDA from binding and activating EDAR at close to stoichiometric ratios in in vitro binding and activity assays. The antibodies block EDA1 and EDA2 of both mammalian and avian origin and, in vivo, suppress the ability of recombinant Fc-EDA1 to rescue ectodermal dysplasia in Eda-deficient Tabby mice. Moreover, administration of EDA blocking antibodies to pregnant wild type mice induced in developing wild type fetuses a marked and permanent ectodermal dysplasia. These function-blocking anti-EDA antibodies with wide cross-species reactivity will enable study of the developmental and postdevelopmental roles of EDA in a variety of organisms and open the route to therapeutic intervention in conditions in which EDA may be implicated.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/toxicidad , Anticuerpos Neutralizantes/toxicidad , Autoanticuerpos/toxicidad , Displasia Ectodérmica/inducido químicamente , Displasia Ectodérmica/inmunología , Ectodisplasinas/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales de Origen Murino/genética , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/inmunología , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Secuencia de Bases , Bovinos , Línea Celular , Perros , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , Displasia Ectodérmica/patología , Ectodisplasinas/genética , Ectodisplasinas/inmunología , Ectodisplasinas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Embarazo
15.
J Exp Med ; 210(11): 2465-76, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24062413

RESUMEN

NOD2 functions as an intracellular sensor for microbial pathogen and plays an important role in epithelial defense. The loss-of-function mutation of NOD2 is strongly associated with human Crohn's disease (CD). However, the mechanisms of how NOD2 maintains the intestinal homeostasis and regulates the susceptibility of CD are still unclear. Here we found that the numbers of intestinal intraepithelial lymphocytes (IELs) were reduced significantly in Nod2(-/-) mice and the residual IELs displayed reduced proliferation and increased apoptosis. Further study showed that NOD2 signaling maintained IELs via recognition of gut microbiota and IL-15 production. Notably, recovery of IELs by adoptive transfer could reduce the susceptibility of Nod2(-/-) mice to the 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis. Our results demonstrate that recognition of gut microbiota by NOD2 is important to maintain the homeostasis of IELs and provide a clue that may link NOD2 variation to the impaired innate immunity and higher susceptibility in CD.


Asunto(s)
Epitelio/inmunología , Homeostasis , Intestinos/inmunología , Intestinos/microbiología , Linfocitos/metabolismo , Microbiota , Proteína Adaptadora de Señalización NOD2/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Suplementos Dietéticos , Susceptibilidad a Enfermedades , Epitelio/efectos de los fármacos , Epitelio/patología , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Interleucina-15/metabolismo , Intestinos/efectos de los fármacos , Intestinos/patología , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal/efectos de los fármacos , Bazo/citología , Timo/citología , Ácido Trinitrobencenosulfónico
16.
J Exp Med ; 210(10): 1899-910, 2013 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-23980097

RESUMEN

The success of allogeneic hematopoietic cell transplantation is limited by acute graft-versus-host disease (GvHD), a severe complication accompanied by high mortality rates. Yet, the molecular mechanisms initiating this disease remain poorly defined. In this study, we show that, after conditioning therapy, intestinal commensal bacteria and the damage-associated molecular pattern uric acid contribute to Nlrp3 inflammasome-mediated IL-1ß production and that gastrointestinal decontamination and uric acid depletion reduced GvHD severity. Early blockade of IL-1ß or genetic deficiency of the IL-1 receptor in dendritic cells (DCs) and T cells improved survival. The Nlrp3 inflammasome components Nlrp3 and Asc, which are required for pro-IL-1ß cleavage, were critical for the full manifestation of GvHD. In transplanted mice, IL-1ß originated from multiple intestinal cell compartments and exerted its effects on DCs and T cells, the latter being preferentially skewed toward Th17. Compatible with these mouse data, increased levels of active caspase-1 and IL-1ß were found in circulating leukocytes and intestinal GvHD lesions of patients. Thus, the identification of a crucial role for the Nlrp3 inflammasome sheds new light on the pathogenesis of GvHD and opens a potential new avenue for the targeted therapy of this severe complication.


Asunto(s)
Proteínas Portadoras/metabolismo , Enfermedad Injerto contra Huésped/etiología , Inflamasomas/metabolismo , Enfermedad Aguda , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Caspasa 1/metabolismo , Proteínas del Citoesqueleto/deficiencia , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Interleucina-17/biosíntesis , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Trasplante Homólogo , Factores de Necrosis Tumoral/biosíntesis
17.
Immunity ; 38(6): 1154-63, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23809162

RESUMEN

Omega-3 fatty acids (ω-3 FAs) have potential anti-inflammatory activity in a variety of inflammatory human diseases, but the mechanisms remain poorly understood. Here we show that stimulation of macrophages with ω-3 FAs, including eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and other family members, abolished NLRP3 inflammasome activation and inhibited subsequent caspase-1 activation and IL-1ß secretion. In addition, G protein-coupled receptor 120 (GPR120) and GPR40 and their downstream scaffold protein ß-arrestin-2 were shown to be involved in inflammasome inhibition induced by ω-3 FAs. Importantly, ω-3 FAs also prevented NLRP3 inflammasome-dependent inflammation and metabolic disorder in a high-fat-diet-induced type 2 diabetes model. Our results reveal a mechanism through which ω-3 FAs repress inflammation and prevent inflammation-driven diseases and suggest the potential clinical use of ω-3 FAs in gout, autoinflammatory syndromes, or other NLRP3 inflammasome-driven inflammatory diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Inflamasomas/metabolismo , Inflamación/prevención & control , Macrófagos/efectos de los fármacos , Animales , Arrestinas/metabolismo , Proteínas Portadoras/genética , Caspasa 1/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/inmunología , Dieta Alta en Grasa/efectos adversos , Activación Enzimática/efectos de los fármacos , Ácidos Grasos Omega-3/inmunología , Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Acoplados a Proteínas G/metabolismo , Arrestina beta 2 , beta-Arrestinas
18.
PLoS One ; 8(4): e61350, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23620746

RESUMEN

Myeloid cells express the TNF family ligands BAFF/BLyS and APRIL, which exert their effects on B cells at different stages of differentiation via the receptors BAFFR, TACI (Transmembrane Activator and CAML-Interactor) and/or BCMA (B Cell Maturation Antigen). BAFF and APRIL are proteins expressed at the cell membrane, with both extracellular and intracellular domains. Therefore, receptor/ligand engagement may also result in signals in ligand-expressing cells via so-called "reverse signalling". In order to understand how TACI-Fc (atacicept) technically may mediate immune stimulation instead of suppression, we investigated its potential to activate reverse signalling through BAFF and APRIL. BAFFR-Fc and TACI-Fc, but not Fn14-Fc, reproducibly stimulated the ERK and other signalling pathways in bone marrow-derived mouse macrophages. However, these effects were independent of BAFF or APRIL since the same activation profile was observed with BAFF- or APRIL-deficient cells. Instead, cell activation correlated with the presence of high molecular mass forms of BAFFR-Fc and TACI-Fc and was strongly impaired in macrophages deficient for Fc receptor gamma chain. Moreover, a TACI-Fc defective for Fc receptor binding elicited no detectable signal. Although these results do not formally rule out the existence of BAFF or APRIL reverse signalling (via pathways not tested in this study), they provide no evidence in support of reverse signalling and point to the importance of using appropriate specificity controls when working with Fc receptor-expressing myeloid cells.


Asunto(s)
Factor Activador de Células B/metabolismo , Membrana Celular/metabolismo , Células Mieloides/citología , Células Mieloides/metabolismo , Transducción de Señal , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Receptor del Factor Activador de Células B/metabolismo , Cromatografía en Gel , Células HEK293 , Humanos , Macrófagos/metabolismo , Ratones , Receptores Fc/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Solubilidad , Receptor de TWEAK
19.
Proc Natl Acad Sci U S A ; 109(45): 18384-9, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-23090995

RESUMEN

A chronic inflammatory microenvironment favors tumor progression through molecular mechanisms that are still incompletely defined. In inflammation-induced skin cancers, IL-1 receptor- or caspase-1-deficient mice, or mice specifically deficient for the inflammasome adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD) in myeloid cells, had reduced tumor incidence, pointing to a role for IL-1 signaling and inflammasome activation in tumor development. However, mice fully deficient for ASC were not protected, and mice specifically deficient for ASC in keratinocytes developed more tumors than controls, suggesting that, in contrast to its proinflammatory role in myeloid cells, ASC acts as a tumor-suppressor in keratinocytes. Accordingly, ASC protein expression was lost in human cutaneous squamous cell carcinoma, but not in psoriatic skin lesions. Stimulation of primary mouse keratinocytes or the human keratinocyte cell line HaCaT with UVB induced an ASC-dependent phosphorylation of p53 and expression of p53 target genes. In HaCaT cells, ASC interacted with p53 at the endogenous level upon UVB irradiation. Thus, ASC in different tissues may influence tumor growth in opposite directions: it has a proinflammatory role in infiltrating cells that favors tumor development, but it also limits keratinocyte proliferation in response to noxious stimuli, possibly through p53 activation, which helps suppressing tumors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Epitelio/patología , Inflamasomas/metabolismo , Neoplasias Cutáneas/patología , Piel/patología , 9,10-Dimetil-1,2-benzantraceno , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/deficiencia , Caspasa 1/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/patología , Citocinas/biosíntesis , Proteínas del Citoesqueleto/deficiencia , Regulación hacia Abajo , Epitelio/metabolismo , Humanos , Inflamación/patología , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Ratones Noqueados , Células Mieloides/metabolismo , Células Mieloides/patología , Neoplasias de Células Escamosas/patología , Especificidad de Órganos , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/prevención & control , Acetato de Tetradecanoilforbol , Microambiente Tumoral , Proteína p53 Supresora de Tumor/metabolismo
20.
J Immunol ; 188(8): 3820-8, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22412192

RESUMEN

Nucleotide-binding oligomerization domain-like receptors (NLRs) are intracellular proteins involved in innate-driven inflammatory responses. The function of the family member NLR caspase recruitment domain containing protein 5 (NLRC5) remains a matter of debate, particularly with respect to NF-κB activation, type I IFN, and MHC I expression. To address the role of NLRC5, we generated Nlrc5-deficient mice (Nlrc5(Δ/Δ)). In this article we show that these animals exhibit slightly decreased CD8(+) T cell percentages, a phenotype compatible with deregulated MHC I expression. Of interest, NLRC5 ablation only mildly affected MHC I expression on APCs and, accordingly, Nlrc5(Δ/Δ) macrophages efficiently primed CD8(+) T cells. In contrast, NLRC5 deficiency dramatically impaired basal expression of MHC I in T, NKT, and NK lymphocytes. NLRC5 was sufficient to induce MHC I expression in a human lymphoid cell line, requiring both caspase recruitment and LRR domains. Moreover, endogenous NLRC5 localized to the nucleus and occupied the proximal promoter region of H-2 genes. Consistent with downregulated MHC I expression, the elimination of Nlrc5(Δ/Δ) lymphocytes by cytotoxic T cells was markedly reduced and, in addition, we observed low NLRC5 expression in several murine and human lymphoid-derived tumor cell lines. Hence, loss of NLRC5 expression represents an advantage for evading CD8(+) T cell-mediated elimination by downmodulation of MHC I levels-a mechanism that may be exploited by transformed cells. Our data show that NLRC5 acts as a key transcriptional regulator of MHC I in lymphocytes and support an essential role for NLRs in directing not only innate but also adaptive immune responses.


Asunto(s)
Genes MHC Clase I , Péptidos y Proteínas de Señalización Intracelular/inmunología , Linfocitos T Citotóxicos/inmunología , Inmunidad Adaptativa , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Médula Ósea/inmunología , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/inmunología , Proliferación Celular , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/genética , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , Linfocitos T Citotóxicos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...