Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Rep ; 36(12): 109748, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34551300

RESUMEN

Obesity-induced inflammation is a major driving force in the development of insulin resistance, type 2 diabetes (T2D), and related metabolic disorders. During obesity, macrophages accumulate in the visceral adipose tissue, creating a low-grade inflammatory environment. Nuclear factor κB (NF-κB) signaling is a central coordinator of inflammatory responses and is tightly regulated by the anti-inflammatory protein A20. Here, we find that myeloid-specific A20-deficient mice are protected from diet-induced obesity and insulin resistance despite an inflammatory environment in their metabolic tissues. Macrophages lacking A20 show impaired mitochondrial respiratory function and metabolize more palmitate both in vitro and in vivo. We hypothesize that A20-deficient macrophages rely more on palmitate oxidation and metabolize the fat present in the diet, resulting in a lean phenotype and protection from metabolic disease. These findings reveal a role for A20 in regulating macrophage immunometabolism.


Asunto(s)
Ácidos Grasos/metabolismo , Obesidad/patología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Tejido Adiposo Blanco/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hidroliasas/genética , Hidroliasas/metabolismo , Resistencia a la Insulina , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Obesidad/metabolismo , Consumo de Oxígeno , Palmitatos/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/deficiencia , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
2.
Cell Death Differ ; 28(5): 1532-1547, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33262469

RESUMEN

Ileal epithelial cell apoptosis and the local microbiota modulate the effects of oxaliplatin against proximal colon cancer by modulating tumor immunosurveillance. Here, we identified an ileal immune profile associated with the prognosis of colon cancer and responses to chemotherapy. The whole immune ileal transcriptome was upregulated in poor-prognosis patients with proximal colon cancer, while the colonic immunity of healthy and neoplastic areas was downregulated (except for the Th17 fingerprint) in such patients. Similar observations were made across experimental models of implanted and spontaneous murine colon cancer, showing a relationship between carcinogenesis and ileal inflammation. Conversely, oxaliplatin-based chemotherapy could restore a favorable, attenuated ileal immune fingerprint in responders. These results suggest that chemotherapy inversely shapes the immune profile of the ileum-tumor axis, influencing clinical outcome.


Asunto(s)
Neoplasias del Colon/fisiopatología , Enfermedades del Íleon/complicaciones , Íleon/patología , Animales , Humanos , Ratones , Pronóstico
3.
Nat Immunol ; 21(4): 381-387, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205881

RESUMEN

Protein ubiquitination regulates protein stability and modulates the composition of signaling complexes. A20 is a negative regulator of inflammatory signaling, but the molecular mechanisms involved are ill understood. Here, we generated Tnfaip3 gene-targeted A20 mutant mice bearing inactivating mutations in the zinc finger 7 (ZnF7) and ZnF4 ubiquitin-binding domains, revealing that binding to polyubiquitin is essential for A20 to suppress inflammatory disease. We demonstrate that a functional ZnF7 domain was required for recruiting A20 to the tumor necrosis factor receptor 1 (TNFR1) signaling complex and to suppress inflammatory signaling and cell death. The combined inactivation of ZnF4 and ZnF7 phenocopied the postnatal lethality and severe multiorgan inflammation of A20-deficient mice. Conditional tissue-specific expression of mutant A20 further revealed the key role of ubiquitin-binding in myeloid and intestinal epithelial cells. Collectively, these results demonstrate that the anti-inflammatory and cytoprotective functions of A20 are largely dependent on its ubiquitin-binding properties.


Asunto(s)
Inflamación/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Epiteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Poliubiquitina/metabolismo , Unión Proteica/fisiología , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Dedos de Zinc/fisiología
4.
Cell Death Dis ; 11(1): 60, 2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974345

RESUMEN

The original version of this article contained an error in the name of one of the co-authors (Wim Declercq). This has been corrected in the PDF and HTML versions.

5.
Nat Cancer ; 1(6): 620-634, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-35121975

RESUMEN

Colorectal cancer (CRC) is highly prevalent in Western society, and increasing evidence indicates strong contributions of environmental factors and the intestinal microbiota to CRC initiation, progression and even metastasis. We have identified a synergistic inflammatory tumor-promoting mechanism through which the resident intestinal microbiota boosts invasive CRC development in an epithelial-to-mesenchymal transition-prone tissue environment. Intestinal epithelial cell (IEC)-specific transgenic expression of the epithelial-to-mesenchymal transition regulator Zeb2 in mice (Zeb2IEC-Tg/+) leads to increased intestinal permeability, myeloid cell-driven inflammation and spontaneous invasive CRC development. Zeb2IEC-Tg/+ mice develop a dysplastic colonic epithelium, which progresses to severely inflamed neoplastic lesions while the small intestinal epithelium remains normal. Zeb2IEC-Tg/+ mice are characterized by intestinal dysbiosis, and microbiota depletion with broad-spectrum antibiotics or germ-free rederivation completely prevents cancer development. Zeb2IEC-Tg/+ mice represent the first mouse model of spontaneous microbiota-dependent invasive CRC and will help us to better understand host-microbiome interactions driving CRC development in humans.


Asunto(s)
Carcinoma , Microbiota , Animales , Carcinoma/metabolismo , Colon/metabolismo , Ratones
7.
Cell Death Dis ; 10(10): 692, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31534131

RESUMEN

The cytokine TNF promotes inflammation either directly by activating the MAPK and NF-κB signaling pathways, or indirectly by triggering cell death. A20 is a potent anti-inflammatory molecule, and mutations in the gene encoding A20 are associated with a wide panel of inflammatory pathologies, both in human and in the mouse. Binding of TNF to TNFR1 triggers the NF-κB-dependent expression of A20 as part of a negative feedback mechanism preventing sustained NF-κB activation. Apart from acting as an NF-κB inhibitor, A20 is also well-known for its ability to counteract the cytotoxic potential of TNF. However, the mechanism by which A20 mediates this function and the exact cell death modality that it represses have remained incompletely understood. In the present study, we provide in vitro and in vivo evidences that deletion of A20 induces RIPK1 kinase-dependent and -independent apoptosis upon single TNF stimulation. We show that constitutively expressed A20 is recruited to TNFR1 signaling complex (Complex I) via its seventh zinc finger (ZF7) domain, in a cIAP1/2-dependent manner, within minutes after TNF sensing. We demonstrate that Complex I-recruited A20 protects cells from apoptosis by stabilizing the linear (M1) ubiquitin network associated to Complex I, a process independent of its E3 ubiquitin ligase and deubiquitylase (DUB) activities and which is counteracted by the DUB CYLD, both in vitro and in vivo. In absence of linear ubiquitylation, A20 is still recruited to Complex I via its ZF4 and ZF7 domains, but this time protects the cells from death by deploying its DUB activity. Together, our results therefore demonstrate two distinct molecular mechanisms by which constitutively expressed A20 protect cells from TNF-induced apoptosis.


Asunto(s)
Receptores Tipo I de Factores de Necrosis Tumoral/efectos adversos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/uso terapéutico , Ubiquitina/efectos de los fármacos , Animales , Apoptosis , Humanos , Ratones , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología
8.
Autophagy ; 15(9): 1657-1659, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31184523

RESUMEN

The C-terminal domain of ATG16L1 includes 7 WD40-type repeats (WD40 domain, WDD) and is not required for canonical macroautophagy/autophagy. Instead, the WDD allows ATG16L1 to induce LC3/Atg8 lipidation in single-membrane compartments, although a detailed functional characterization of this region is still missing. In a recent report we identify the anti-inflammatory molecule TNFAIP3/A20 as a binding partner of the WDD. Such physical interaction allows mutual downregulation of the expression levels of both proteins, so that the absence of one of them causes upregulation of the other. This cross-regulation provides a molecular basis for a striking genetic interaction in mice where elimination of both molecules in the intestinal epithelium generates an aggressive inflammatory phenotype. In vitro studies reveal unexpected features of the functional interplay between ATG16L1 and TNFAIP3. ATG16L1 requires TNFAIP3 to sustain the canonical autophagic flux measured by SQSTM1/p62 degradation. The WDD mediates lysosomal degradation of TNFAIP3 promoted by ATG16L1, and also regulates the NFKB/NF-κB response. Therefore, our data reveal new roles of the WDD and TNFAIP3 in the regulation of autophagy, protein stability and inflammatory signaling. More generally, we identify the interaction between ATG16L1 and TNFAIP3 as a signaling hub that integrates different pathways with important implications for intestinal homeostasis.


Asunto(s)
Autofagia , Animales , Antiinflamatorios , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras , Homeostasis , Ratones , FN-kappa B , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
9.
Nat Commun ; 10(1): 1834, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015422

RESUMEN

Prevention of inflammatory bowel disease (IBD) relies on tight control of inflammatory, cell death and autophagic mechanisms, but how these pathways are integrated at the molecular level is still unclear. Here we show that the anti-inflammatory protein A20 and the critical autophagic mediator Atg16l1 physically interact and synergize to regulate the stability of the intestinal epithelial barrier. A proteomic screen using the WD40 domain of ATG16L1 (WDD) identified A20 as a WDD-interacting protein. Loss of A20 and Atg16l1 in mouse intestinal epithelium induces spontaneous IBD-like pathology, as characterized by severe inflammation and increased intestinal epithelial cell death in both small and large intestine. Mechanistically, absence of A20 promotes Atg16l1 accumulation, while elimination of Atg16l1 or expression of WDD-deficient Atg16l1 stabilizes A20. Collectively our data show that A20 and Atg16l1 cooperatively control intestinal homeostasis by acting at the intersection of inflammatory, autophagy and cell death pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Repeticiones WD40/genética , Animales , Autofagia/inmunología , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Endoscopía , Femenino , Homeostasis/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/diagnóstico por imagen , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/citología , Mucosa Intestinal/diagnóstico por imagen , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica/inmunología , Proteómica , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Repeticiones WD40/inmunología
10.
Front Immunol ; 9: 769, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29692786

RESUMEN

Optineurin (OPTN) was identified 20 years ago in a yeast-two-hybrid screen with a viral protein known to inhibit the cytolytic effects of tumor necrosis factor. Since then, OPTN has been identified as a ubiquitin-binding protein involved in many signaling pathways and cellular processes, and mutations in the OPTN gene have been associated with glaucoma, Paget's disease of bone and neurodegenerative pathologies. Its role in autophagy, however, has attracted most attention in recent years and may explain (some of) the mechanisms behind the disease-associated mutations of OPTN. In this brief review, we focus on the role of OPTN in inflammation and immunity and describe how this may translate to its involvement in human disease.


Asunto(s)
Inmunidad/inmunología , Transducción de Señal/inmunología , Factor de Transcripción TFIIIA/inmunología , Animales , Proteínas de Ciclo Celular , Humanos , Proteínas de Transporte de Membrana
11.
Trends Immunol ; 37(9): 621-633, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27480243

RESUMEN

Optineurin (OPTN) was initially identified as a regulator of NF-κB and interferon signaling, but attracted most attention because of its association with various human disorders such as glaucoma, Paget disease of bone, and amyotrophic lateral sclerosis. Importantly, OPTN has recently been identified as an autophagy receptor important for the autophagic removal of pathogens, damaged mitochondria, and protein aggregates. This activity is most likely compromised in patients carrying OPTN mutations, and contributes to the observed phenotypes. In this review we summarize recent studies describing the molecular mechanisms by which OPTN controls immunity and autophagy, and discuss these findings in the context of several diseases that have been associated with OPTN (mal)function.


Asunto(s)
Esclerosis Amiotrófica Lateral/inmunología , Autofagia/genética , Glaucoma/inmunología , Osteítis Deformante/inmunología , Factor de Transcripción TFIIIA/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Proteínas de Ciclo Celular , Glaucoma/genética , Humanos , Inmunidad/genética , Interferones/metabolismo , Proteínas de Transporte de Membrana , Mutación/genética , FN-kappa B/metabolismo , Osteítis Deformante/genética , Transducción de Señal , Factor de Transcripción TFIIIA/genética
12.
Eur J Immunol ; 46(4): 971-80, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26677802

RESUMEN

Optineurin (OPTN) is an evolutionary conserved and ubiquitously expressed ubiquitin-binding protein that has been implicated in glaucoma, Paget bone disease, amyotrophic lateral sclerosis, and other neurodegenerative diseases. From in vitro studies, OPTN was shown to suppress TNF-induced NF-κB signaling and virus-induced IRF signaling, and was identified as an autophagy receptor required for the clearance of cytosolic Salmonella upon infection. To assess the in vivo functions of OPTN in inflammation and infection, we generated OPTN-deficient mice. OPTN knockout mice are born with normal Mendelian distribution and develop normally without any signs of spontaneous organ abnormality or inflammation. However, no differences in NF-κB activation could be observed in OPTN knockout mice or fibroblasts derived from these mice upon TNF or LPS treatment. Primary bone marrow-derived macrophages from OPTN-deficient mice had slightly impaired IRF signaling and reduced IFN type I production in response to LPS or poly(I,C). Finally, OPTN-deficient mice were more susceptible to infection with Salmonella, confirming in vivo the importance of OPTN in bacterial clearance.


Asunto(s)
Proteínas del Ojo/genética , FN-kappa B/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Proteínas de Ciclo Celular , Fibroblastos/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Poli I-C/farmacología , Infecciones por Salmonella/microbiología , Transducción de Señal/inmunología
13.
Nat Commun ; 6: 5794, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25565005

RESUMEN

Early T-cell precursor leukaemia (ETP-ALL) is a high-risk subtype of human leukaemia that is poorly understood at the molecular level. Here we report translocations targeting the zinc finger E-box-binding transcription factor ZEB2 as a recurrent genetic lesion in immature/ETP-ALL. Using a conditional gain-of-function mouse model, we demonstrate that sustained Zeb2 expression initiates T-cell leukaemia. Moreover, Zeb2-driven mouse leukaemia exhibit some features of the human immature/ETP-ALL gene expression signature, as well as an enhanced leukaemia-initiation potential and activated Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signalling through transcriptional activation of IL7R. This study reveals ZEB2 as an oncogene in the biology of immature/ETP-ALL and paves the way towards pre-clinical studies of novel compounds for the treatment of this aggressive subtype of human T-ALL using our Zeb2-driven mouse model.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Leucemia de Células T/fisiopatología , Proteínas Represoras/genética , Transducción de Señal/fisiología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Técnicas Histológicas , Proteínas de Homeodominio/inmunología , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Quinasas Janus/metabolismo , Estimación de Kaplan-Meier , Cariotipificación , Luciferasas , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-7/metabolismo , Proteínas Represoras/inmunología , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
14.
Nat Commun ; 5: 5103, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25267258

RESUMEN

The transcription factor NF-κB is indispensable for intestinal immune homeostasis, but contributes to chronic inflammation and inflammatory bowel disease (IBD). A20, an inhibitor of both NF-κB and apoptotic signalling, was identified as a susceptibility gene for multiple inflammatory diseases, including IBD. Despite absence of spontaneous intestinal inflammation in intestinal epithelial cell (IEC) specific A20 knockout mice, we found additional myeloid-specific A20 deletion to synergistically drive intestinal pathology through cell-specific mechanisms. A20 ensures intestinal barrier stability by preventing cytokine-induced IEC apoptosis, while A20 prevents excessive cytokine production in myeloid cells. Combining IEC and myeloid A20 deletion induces ileitis and severe colitis, characterized by IEC apoptosis, Paneth and goblet cell loss, epithelial hyperproliferation and intestinal microbiota dysbiosis. Continuous epithelial cell death and regeneration in an inflammatory environment sensitizes cells for neoplastic transformation and the development of colorectal tumours in aged mice.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Células Epiteliales/enzimología , Intestinos/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Apoptosis , Colitis/enzimología , Colitis/genética , Colitis/patología , Colitis/fisiopatología , Cisteína Endopeptidasas/genética , Células Epiteliales/citología , Células Epiteliales/patología , Femenino , Células Caliciformes/citología , Células Caliciformes/enzimología , Células Caliciformes/patología , Homeostasis , Humanos , Intestinos/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células de Paneth/citología , Células de Paneth/enzimología , Células de Paneth/patología , Especificidad de la Especie , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
15.
PLoS One ; 8(6): e67855, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840778

RESUMEN

The functions of actin family members during development are poorly understood. To investigate the role of beta-actin in mammalian development, a beta-actin knockout mouse model was used. Homozygous beta-actin knockout mice are lethal at embryonic day (E)10.5. At E10.25 beta-actin knockout embryos are growth retarded and display a pale yolk sac and embryo proper that is suggestive of altered erythropoiesis. Here we report that lack of beta-actin resulted in a block of primitive and definitive hematopoietic development. Reduced levels of Gata2, were associated to this phenotype. Consistently, ChIP analysis revealed multiple binding sites for beta-actin in the Gata2 promoter. Gata2 mRNA levels were almost completely rescued by expression of an erythroid lineage restricted ROSA26-promotor based GATA2 transgene. As a result, erythroid differentiation was restored and the knockout embryos showed significant improvement in yolk sac and embryo vascularization. These results provide new molecular insights for a novel function of beta-actin in erythropoiesis by modulating the expression levels of Gata2 in vivo.


Asunto(s)
Actinas/fisiología , Desarrollo Embrionario , Células Precursoras Eritroides/citología , Eritropoyesis/fisiología , Factor de Transcripción GATA2/metabolismo , Células Madre Hematopoyéticas/citología , Animales , Circulación Sanguínea , Células Cultivadas , Células Precursoras Eritroides/metabolismo , Femenino , Factor de Transcripción GATA2/genética , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA